US20190172588A1 - Pharmacogenetic drug interaction management system - Google Patents

Pharmacogenetic drug interaction management system Download PDF

Info

Publication number
US20190172588A1
US20190172588A1 US16/267,651 US201916267651A US2019172588A1 US 20190172588 A1 US20190172588 A1 US 20190172588A1 US 201916267651 A US201916267651 A US 201916267651A US 2019172588 A1 US2019172588 A1 US 2019172588A1
Authority
US
United States
Prior art keywords
drug
gene
genetic
interactions
clinical
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/267,651
Inventor
Bao Tran
Ha Tran
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/843,903 external-priority patent/US20140278475A1/en
Application filed by Individual filed Critical Individual
Priority to US16/267,651 priority Critical patent/US20190172588A1/en
Publication of US20190172588A1 publication Critical patent/US20190172588A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/30ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for calculating health indices; for individual health risk assessment
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06NCOMPUTING ARRANGEMENTS BASED ON SPECIFIC COMPUTATIONAL MODELS
    • G06N20/00Machine learning
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H15/00ICT specially adapted for medical reports, e.g. generation or transmission thereof
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H70/00ICT specially adapted for the handling or processing of medical references
    • G16H70/40ICT specially adapted for the handling or processing of medical references relating to drugs, e.g. their side effects or intended usage
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06NCOMPUTING ARRANGEMENTS BASED ON SPECIFIC COMPUTATIONAL MODELS
    • G06N3/00Computing arrangements based on biological models
    • G06N3/02Neural networks
    • G06N3/04Architecture, e.g. interconnection topology
    • G06N3/045Combinations of networks
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06NCOMPUTING ARRANGEMENTS BASED ON SPECIFIC COMPUTATIONAL MODELS
    • G06N7/00Computing arrangements based on specific mathematical models
    • G06N7/01Probabilistic graphical models, e.g. probabilistic networks

Definitions

  • the present invention relates to the field of computerized drug identification.
  • Drug interactions remain a principal aspect of the pharmaceutical sciences.
  • a drug interaction is a commonly known situation in which a substance affects the activity of a drug, such that the effects of a given drug is increased or decreased, or the combination of the substance and the drug produce a new effect that neither produces alone.
  • drug-drug interactions are most unpredictable; however, drug-food interactions also are known to exist between drugs and foods, as well as drug-herb interactions between drugs and herbs.
  • a system includes code to match genomic biomarker(s) from gene or DNA sequencing for a population with historical information for a population on drug structure, dosage, clinical variability and risk for adverse events for the drug substance, the computer constructing side effect features for each drug, and a classifier to predict whether a drug pair results in adverse interactions, the computer generating one or more indicia for the drug substance; and code to apply the indicia to select a custom dosage for a drug whose properties are selected and crafted for the individual patient to provide personalized medicine.
  • a system includes code to: acquire subject genetic scans using a gene sequencer; identify each substance to be provided to the subject; determine substance interactions for each drug and the genetic scan; and provide indicia associated with each substance to warn the subject or a medical provider based on the genetic scan.
  • a system in another aspect, includes a substance to be consumed by a subject and one or more indicia labeling the substance with: genomic biomarkers; drug exposure and clinical response variability; risk for adverse events; genotype-specific dosing; polymorphic drug target and disposition genes; and treatment based on the biomarker.
  • the system may make medical trials more efficient. This will lower the costs that come about due to adverse drug side effects and prescription of drugs that have been proven ineffective in certain genotypes. Drug companies can develop and license a drug specifically intended for those who are the small population genetically at risk for adverse side effects.
  • FIGS. 1A-1B show exemplary substance interaction management processes
  • FIG. 2 is a schematic illustration of a data processing system configured for computer management genetic information, precision medication and drug interaction information retrieval;
  • FIG. 3 is a flow chart illustrating a process for pharmacogenetics drug interaction information retrieval.
  • FIG. 4 shows a big data learning machine to process genetic data and determine pharmacogenetics relationship among genes and drugs for drug interaction purposes.
  • FIG. 5A shows various common aberrations in cancer genomes.
  • FIG. 5B shows an exemplary system to detect the evolutionary paths of escape.
  • FIG. 5C shows an exemplary model generated by the system of FIG. 5B .
  • FIG. 5D shows an exemplary a heterogeneous collection of normal cells and cancer subclones developed during an evolutionary history of a tumor.
  • Embodiments of the present invention provide a method, system and computer program product for computer identification (scanning or imaging of drugs) for drug interaction information retrieval.
  • multiple different drugs can be scanned or imaged to detect identifiable content disposed on the different drugs.
  • Each drug can be compared to a data store of drug information to identify each drug.
  • pharmacogenetics data and drug interaction data can be retrieved for each identified drug.
  • known drug-drug interactions and genetic impacts for the identified drugs can be determined and a report can be provided to include the known drug-drug interactions. In this way, precision medicine and drug-drug interactions resulting from the use of the multiple different drugs can be determined without recourse to a voluminous text of drug interactions.
  • FIGS. 1A-1B show exemplary processes for computer detection of substances or drugs for drug interaction information retrieval.
  • the process cryptographically reads substance content from RF tag or barcode on a secure bottle and in 20 identifies content for the substance.
  • identified substances are added to an interaction list and in 40 the process determines if additional substances remain to be scanned (RF/Bar Code), and if so loops back to 10 .
  • the interaction list now populated by a list of scanned substances is processed.
  • the process retrieves drug data and drug interaction data for each of the scanned substances and determines relative interactions between substances.
  • the process receives genetic scans for subjects.
  • the process applies pharmacogenomic information to the drug-gene interaction data and selects the best medication and identify people who need an unusually high or low dose.
  • relative interactions can be rendered within a report such as a paper report or a graphical user interface display.
  • FIG. 1B a camera based drug identification and drug interaction management system is shown.
  • images of a substance such as a drug can be acquired.
  • the process identifies content for the substance can be retrieved from the image.
  • substances can be identified according to the identifying content.
  • the identified substance can be added to an interaction list.
  • the process loops back to 110 if additional substances remain to be imaged and if not, the interaction list now populated by a list of imaged substances can be processed.
  • the process receives genetic scans for subjects.
  • the process retrieves drug data and drug interaction data for each of the imaged substances and determines relative interactions between substances, and in 180 pharmacogenomic information is applied to select the best medication and identify people who need an unusually high or low dose.
  • the relative interactions can be rendered within a report such as a paper report or a graphical user interface display.
  • FIG. 2 schematically depicts a data processing system configured for computer visualization of drugs for drug interaction information retrieval.
  • the system can include a host computing platform 202 coupled to a camera 220 such as a digital still camera or digital video camera.
  • the camera 220 can be focused on a marshalling point 240 provided by a marshalling apparatus 230 , for example gravity feed or isolation chamber or miniature conveyor belt.
  • the host computing platform 202 also can be communicatively coupled a drug image data store 250 of known substances and corresponding known identifying content visually disposed on the known substances.
  • the host computing platform 202 additionally can be communicatively coupled to a drug interaction data store 260 providing drug interaction data for different substances relative to other substances including prescription and over-the-counter drugs, vitamins and herbal remedies, and food products.
  • multiple different substances such as prescription drugs, over-the-counter drugs or even vitamins and herbal remedies can be provided to a marshalling apparatus such as a gravity feed or miniature conveyor belt or even a chamber.
  • the marshalling apparatus can isolate an individual one of the different substances for imaging by camera 220 , for example a charge coupled device (CCD) driven digital camera or video recorder.
  • the camera 220 can capture an image of each individual one of the different substances 110 A, 110 B, 110 N and computer visualization for drug interaction information retrieval logic 300 can process each captured image to detect identifying content disposed on each of the different substances such as a pill marking or code.
  • CCD charge coupled device
  • the computer visualization for drug interaction information retrieval logic 300 in turn can compare the identified content to a data store of known substances 140 to identify each of the different substances.
  • the computer can lookup not only known drug interactions for each of the different substances, but also known drug interactions between the identified ones of the substances and pharmacogenetics impact on the individual patient. Thereafter, a drug interaction report can be produced indicating the known drug interactions between the identified ones of the substances.
  • the host computing platform 202 can support the execution of computer scanning or visualization for drug interaction information retrieval logic 270 .
  • the logic can include program code enabled to acquire imagery of different substances in the marshalling point 240 .
  • the program code further can be enabled to locate and retrieve identifying content disposed on the different substances and to look up the identifying content in the drug image data store 250 in order to identify each of the substances.
  • the program code yet further can be enabled to retrieve from drug interaction data store 260 drug interactions for each of the identified substances and to particularly correlate the retrieved drug interactions to different ones of the substances so that relative drug interactions can be determined for the substances.
  • the program code can be enabled to render a report of drug interaction data in a graphical user interface display 280 of drug interaction data.
  • the computing platform 202 also receives pharmacogenetics interaction 282 .
  • the host computing platform 202 can support the execution of computer visualization for pharmacogenetics interaction information retrieval logic 272 .
  • Genetic information is captured by high speed gene sequencing machine 210 that uploads gene data to a cloud computing network 212 .
  • the doctors, pharmacists, or consumers can access DNA information using mobile computers such as smart phone 214 , for example.
  • the system can have wireless communication 292 with the medication's labels.
  • the labels can have RF tags or NFC tags that provide upon inquiry FDA required labeling contents.
  • the content can be genomic biomarkers; drug exposure and clinical response variability; risk for adverse events; genotype-specific dosing; polymorphic drug target and disposition genes; and treatment based on the biomarker.
  • NFC tags are passive devices and operate without a power supply of their own and are reliant on an active device to come into range before they are activated. To power these NFC tags, electromagnetic induction is used to create a current in the passive device.
  • Active devices such as a reader or a smartphone, are responsible for generating the magnetic field with a simple coil of wire, which produces magnetic fields perpendicular to the flow of the alternating current in the wire.
  • NFC operates over just a few inches, rather than the meters in other types of wireless communication.
  • the system can be used to provide personalized medicine through custom pharmacy compounding 294 or custom production of a drug whose various properties (e.g. dose level, ingredient selection, route of administration, etc.) are selected and crafted for an individual patient (in contrast to mass-produced unit doses or fixed-dose combinations).
  • various properties e.g. dose level, ingredient selection, route of administration, etc.
  • the genetic scan in 70 can be generated by gene sequencing machines.
  • DNA sequencing is the process of determining the precise order of nucleotides within a DNA molecule. It includes any method or technology that is used to determine the order of the four bases—adenine, guanine, cytosine, and thymine—in a strand of DNA.
  • Various high speed sequencers can be used.
  • Nanopore DNA sequencing is based on the readout of electrical signals occurring at nucleotides passing by alpha-hemolysin pores covalently bound with cyclodextrin. The DNA passing through the nanopore changes its ion current.
  • Oxford Nanopore Technologies offers a handheld sequencer capable of generating more than 150 megabases of sequencing data in one run.
  • Another approach uses measurements of the electrical tunnelling currents across single-strand DNA as it moves through a channel. Depending on its electronic structure, each base affects the tunnelling current differently, allowing differentiation between different bases.
  • the use of tunnelling currents has the potential to sequence orders of magnitude faster than ionic current methods and the sequencing of several DNA oligomers and micro-RNA has already been achieved.
  • Sequencing by hybridization is a non-enzymatic method that uses a DNA microarray. A single pool of DNA whose sequence is to be determined is fluorescently labeled and hybridized to an array containing known sequences. Strong hybridization signals from a given spot on the array identify its sequence in the DNA being sequenced. Mass spectrometry may be used to determine DNA sequences.
  • MALDI-TOF MS Matrix-assisted laser desorption ionization time-of-flight mass spectrometry
  • MALDI-TOF MS has specifically been investigated as an alternative method to gel electrophoresis for visualizing DNA fragments.
  • DNA fragments generated by chain-termination sequencing reactions are compared by mass rather than by size.
  • the mass of each nucleotide is different from the others and this difference is detectable by mass spectrometry.
  • Single-nucleotide mutations in a fragment can be more easily detected with MS than by gel electrophoresis alone.
  • MALDI-TOF MS can more easily detect differences between RNA fragments, so researchers may indirectly sequence DNA with MS-based methods by converting it to RNA first.
  • RNAP sequencing is based on use of RNA polymerase (RNAP), which is attached to a polystyrene bead. One end of DNA to be sequenced is attached to another bead, with both beads being placed in optical traps. RNAP motion during transcription brings the beads in closer and their relative distance changes, which can then be recorded at a single nucleotide resolution. The sequence is deduced based on the four readouts with lowered concentrations of each of the four nucleotide types, similarly to the Sanger method. Other high speed gene sequencers can be used.
  • RNAP RNA polymerase
  • the system applies pharmacogenomic information to select the best medication and identify people who need an unusually high or low dose. This is in addition to clinical factors, such as a patient's age, weight, sex, and liver and kidney function.
  • Pharmacogenomics (sometimes called pharmacogenetics) is focused on understanding how genes affect individual responses to medications and to help doctors select the drugs and dosages best suited for each person. Pharmacogenomics looks at variations in genes for proteins that influence drug responses. Such proteins include a number of liver enzymes that convert medications into their active or inactive forms. Even small differences in the genetic sequences of these enzymes can have a big impact on a drug's safety or effectiveness.
  • CYP2D6 a liver enzyme known as CYP2D6.
  • This enzyme acts on a quarter of all prescription drugs, including the painkiller codeine, which it converts into the drug's active form, morphine.
  • the CYP2D6 gene exists in more than 160 different versions, many of which vary by only a single difference in their DNA sequence, although some have larger changes. The majority of these variants don't affect drug responses. Some people have hundreds or even thousands of copies of the CYP2D6 gene (typically, people have two copies of each gene). Those with extra copies of this gene manufacture an overabundance of CYP2D6 enzyme molecules and metabolize the drug very rapidly. As a result, codeine may be converted to morphine so quickly and completely that a standard dose of the drug can be an overdose.
  • CYP2D6 results in a nonfunctional enzyme. People with these variants metabolize codeine slowly, if at all, so they might not experience much pain relief. For these people, doctors might prescribe a different type of pain reliever.
  • Pharmacogenomic information can cover dosage guidance, possible side effects or differences in effectiveness for people with certain genomic variations—can help doctors tailor their drug prescriptions for individual patients.
  • the system applies pharmacogenomic data to develop and market drugs for people with specific genetic profiles. The system can identify the genetic basis for certain serious side effects, drugs could be prescribed only to people who are not at risk for them. As a result, potentially lifesaving medications, which otherwise might be taken off the market because they pose a risk for some people, could still be available to those who could benefit from them. For example, a few drug and gene associations are listed in the Appendix.
  • the computer visualization for drug interaction information retrieval logic 270 is shown to execute in a single host computing platform 202 , the invention is not so limited and the computer visualization for drug interaction information retrieval logic 270 also can be distributed in form across multiple different computing platforms.
  • the camera 220 and marshalling apparatus 230 can be located remotely from the host computing platform 202 whilst providing acquired imagery to the host computing platform 210 over a computer communications network, whether wireless or wirebound.
  • either or both of the drug image data store 250 and the drug interaction data store 260 can be remotely disposed from the host computing platform 202 and accessible over a computer communications network, whether wireless or wirebound.
  • the system enables a medical model that separates patients into different groups—with medical decisions, practices, interventions and/or products being tailored to the individual patient based on their predicted response or risk of disease.
  • Genotyping is the process of obtaining an individual's DNA sequence by using biological assays. By having a detailed account of an individual's DNA sequence, their genome can then be compared to a reference genome, like that of the Human Genome Project, to assess the existing genetic variations that can account for possible diseases.
  • An individual's genetic make-up also plays a large role in how well they respond to a certain treatment, and therefore, knowing their genetic content can change the type of treatment they receive.
  • the system applies pharmacogenomics by using an individual's genome to provide a more informed and tailored drug prescription.
  • personalized medicine can greatly aid the advancements of preventive care. For instance, many women are already being genotyped for certain mutations in the BRCA1 and BRCA2 gene if they are predisposed because of a family history of breast cancer or ovarian cancer. As more causes of diseases are mapped out according to mutations that exist within a genome, the easier they can be identified in an individual. Measures can then be taken to prevent a disease from developing. Even if mutations were found within a genome, having the details of their DNA can reduce the impact or delay the onset of certain diseases. Having the genetic content of an individual will allow better guided decisions in determining the source of the disease and thus treating it or preventing its progression. This will be extremely useful for diseases like Alzheimer's or cancers that are thought to be linked to certain mutations in human DNA.
  • the system can be used to test efficacy and safety of a drug specific to a targeted patient group/sub-group is companion diagnostics.
  • This technology is an assay that is developed during or after a drug is made available on the market and is helpful in enhancing the therapeutic treatment available based on the individual.
  • companion diagnostics have incorporated the pharmacogenomic information related to the drug into their prescription label in an effort to assist in making the most optimal treatment decision possible for the patient.
  • Having an individual's genomic information can be significant in the process of developing drugs as they await approval from the FDA for public use. Having a detailed account of an individual's genetic make-up can be a major asset in deciding if a patient can be chosen for inclusion or exclusion in the final stages of a clinical trial. Being able to identify patients who will benefit most from a clinical trial will increase the safety of patients from adverse outcomes caused by the product in testing, and will allow smaller and faster trials that lead to lower overall costs.
  • drugs that are deemed ineffective for the larger population can gain approval by the FDA by using personal genomes to qualify the effectiveness and need for that specific drug or therapy even though it may only be needed by a small percentage of the population.
  • Treatments can be more specifically tailored to an individual and give insight into how their body will respond to the drug and if that drug will work based on their genome.
  • the personal genotype can allow physicians to have more detailed information that will guide them in their decision in treatment prescriptions, which will be more cost-effective and accurate.
  • FIG. 3 shows a method 300 for predicting drug-drug interactions based on genetic data and clinical side effects, in accordance with an embodiment of the present principles.
  • the process includes the following: At 310 , construct a comprehensive gene-drug-drug interactions (GDDIs) training dataset that includes all pharmaceutical, pharmacokinetic (PK), pharmacogenetic (PG), and pharmacodynamic (PD) GDDIs from multiple data sources for each drug in a set of drugs under consideration.
  • GDDIs gene-drug-drug interactions
  • the multiple data sources can include, but are not limited to, the following: gene sequencers, clinical trials; drug development information; empirical information; a drug bank; drug label information; an adverse event reporting system (e.g., the FDA Adverse Event Reporting System information (FAERS)); and text mining from scientific documents (e.g., search tool for interactions of chemicals (STITCH)).
  • FAERS FDA Adverse Event Reporting System information
  • STITCH search tool for interactions of chemicals
  • the genetic panels are generated by genetic sequencers, and all drugs' side effects, from which the side effect features are constructed, come from one or more of the following sources: clinical trials; drug development; empirical information; FDA drug label (SIDER and DAILYMED®); FDA Adverse Event Reporting System (FAERS); and real-world evidence.
  • the features used for building the classifier can include, but are not limited to, the following: drug's clinical side effect keywords; and other drug properties (e.g., chemical structures, protein targets, and so forth).
  • the term “positive predicted GDDIs” refers to drugs pairs that cannot be taken together given a patient genetic profile.
  • the term “negative predicted GDDIs” refers to drugs pairs that may be safe to use together with a genetic profile.
  • Label side effects means the side effects are recorded in drug labels (for example, but not limited to, SIDER database, DAILYMED®, and so forth).
  • FDA side effects means the side effects are recorded in, for example, but not limited to, the FDA Adverse Event Reporting System (FAERS).
  • FERS FDA Adverse Event Reporting System
  • relative interactions between the different drug substances can be determined by locating references in the interaction data for each of the drug substances to others of the substances.
  • the relative interactions can be rendered within a report such as a paper report or a graphical user interface display.
  • an activatable link can be provided in the display for selected ones of the drug substances for reordering the selected ones of the drug substances.
  • the system can also perform GDDI discovery and prediction that uses molecular structure similarity information derived from fingerprint-based modeling. Identifying new GDDIs using structural similarity is based on the basic idea that if drug A interacts with drug B, and drug C is structurally similar to A, then C should also interact with B (the argument also follows if A is replaced with B). Hence, by combining knowledge of known interactions with structural similarity it is possible to identify new interactions.
  • the process uses a list of drug-drug interactions from DrugBank (step 1), structural similarity computation was carried out using molecular fingerprints (step 2), apply gene-drug interaction to similar drugs, and a new list of gene-drug interactions can be inferred.
  • Structural similarity can be identified in three steps: 1) Collecting and processing drug structures: Information on the structures of the compounds in DrugBank is retrieved along with the SMILE code (a chemical notation representing a chemical structure in linear textual form). 2) Structural representation: BIT_MACCS (MACCS Structural Keys Bit packed) fingerprints are calculated for all molecules included in the study and each molecule is represented as a bit vector that codes the presence or absence of structural features where each feature is assigned a specific bit position. 3) Similarity measures, computation, and data representation: Different measures are used to compare similarity between two molecular fingerprints. In one embodiment, the molecular fingerprints were compared using Tanimoto coefficient (TC).
  • TC Tanimoto coefficient
  • the TC can span values between 0 and 1, where 0 means ‘maximum dissimilarity’ and 1 means ‘maximum similarity.’
  • the TC between two fingerprint representations A and B is defined as the number of features present in the intersection of both fingerprints A and B divided by the number of features present in the union of both fingerprints.
  • the pharmacogenomic information can be applied to drug labeling.
  • One embodiment may contain information on genomic biomarkers and can describe:
  • the information may include specific actions to be taken based on the biomarker information.
  • Pharmacogenomic information can appear in different sections of the labeling depending on the actions.
  • Biomarkers in the table include but are not limited to germ-line or somatic gene variants, functional deficiencies, expression changes, and chromosomal abnormalities; selected protein biomarkers that are used to select patients for treatment are also included.
  • the process includes constructing a gene-drug interactions training dataset that includes pharmaceutical, pharmacokinetic or pharmacodynamics, and pharmacogenomics drug-drug interactions for each drug; constructing side effect features for each of the plurality of drugs from side effects associated with the plurality of drugs; running a gene-drug-drug interactions classifier that predicts adverse drug-drug interactions for drug pairs and the genetic scan; and for each of the side effects, performing a Fisher's exact test to determine predicted gene-drug-drug interactions. Fisher's exact testis a statistical significance test used in the analysis of contingency tables.
  • FIG. 4 shows a deep learning machine using deep convolutionary neural networks for detecting genetic based drug-drug interaction.
  • AlexNet 8-layer architecture
  • VGGNet 16-layer architecture (each pooling layer and last 2 FC layers are applied as feature vector).
  • SIDER a text-mined database of drug package inserts
  • the Offsides database that contains information complementary to that found in SIDER and improves the prediction of protein targets and drug indications
  • Twosides database of mined putative DDIs also lists predicted adverse events, all available at the http://PharmGKB.org Web site.
  • the system of FIG. 4 receives data on adverse events strongly associated with indications for which the indication and the adverse event have a known causative relationship.
  • a drug-event association is synthetic if it has a tight reporting correlation with the indication ( ⁇ 0.1) and a high relative reporting (RR) association score (RR ⁇ 2).
  • Drugs reported frequently with these indications were 80.0 (95% CI, 14.2 to 3132.8; P ⁇ 0.0001, Fisher's exact test) times as likely to have synthetic associations with indication events.
  • Disease indications are a significant source of synthetic associations. The more disproportionately a drug is reported with an indication (x axis), the more likely that drug will be synthetically associated. For example, adverse events strongly associated with drugs are retrieved from the drug's package insert. These drug-event pairs represent a set of known strong positive associations.
  • Adverse events related to sex and race are also analyzed. For example, for physiological reasons, certain events predominantly occur in males (for example, penile swelling and azoospermia). Drugs that are disproportionately reported as causing adverse events in males were more likely to be synthetically associated with these events. Similarly, adverse events that predominantly occur in either relatively young or relatively old patients are analyzed.
  • Off-label adverse event data is also analyzed, and off-label uses refer to any drug effect not already listed on the drug's package insert.
  • SIDER database extracted from drug package inserts, lists 48,577 drug-event associations for 620 drugs and 1092 adverse events that are also covered by the data mining. Offsides recovers 38.8% (18,842 drug-event associations) of SIDER associations from the adverse event reports. Thus, Offsides finds different associations from those reported during clinical trials before drug approval.
  • Twosides Polypharmacy side effects for pairs of drugs (Twosides) are also analyzed. These associations are limited to only those that cannot be clearly attributed to either drug alone (that is, those associations covered in Offsides).
  • the database contains an significant associations for which the drug pair has a higher side-effect association score, determined using the proportional reporting ratio (PRR), than those of the individual drugs alone.
  • PRR proportional reporting ratio
  • the system determines pairwise similarity metrics between all drugs in the Offsides and SIDER databases.
  • the system can predict shared protein targets using drug-effect similarities.
  • the side-effect similarity score between two drugs is linearly related to the number of targets that those drugs share.
  • the system can determine relationships between the proportion of shared indications between a pair of drugs and the similarity of their side-effect profiles in Offsides.
  • the system can use side-effect profiles to suggest new uses for old drugs. While the preferred system predicts existing therapeutic indications of known drugs, the system can recommend drug repurposing using drug-effect similarities in Offsides.
  • the system can identify DDIs shared by an entire drug class.
  • the class-class interaction analysis generates putative drug class interactions.
  • the system analyzes laboratory reports commonly recorded in EMRs that may be used as markers of these class-specific DDIs.
  • the system can be used systematic drug surveillance.
  • the FDA manages a collection of adverse drug event reports to monitor the safety of drugs. They rely on physicians, pharmaceutical companies, and patients to volunteer these reports. Since reporting is not mandatory, many adverse drug events that occur are never reported to the FDA.
  • an embodiment of the present invention uses an algorithm to infer unreported adverse drug events. This embodiment relies on the fact that many adverse events occur together. For example, nausea and vomiting commonly manifest together. Therefore, if a drug is observed to causes nausea, it can be inferred that it also causes vomiting.
  • the processor can analyze evolutions in the target of the treatment and recommend alternative treatment.
  • systems and methods includes analyzing a disease state of a subject by collecting genetic profile data on a population of tumors and original tumor treatment(s); identifying one or more evolutionary paths of escape and evolved tumor treatment(s); and based on a subject profile, predicting a probability of escape along the one or more evolutionary paths.
  • a method for analyzing a disease state of a subject includes capturing a first liquid biopsy from the subject; providing the liquid biopsy to a genetic analyzer to identify the subject's genetic information of a first disease state at a first time point; searching for genetically similar patients and predicting a mutation of the disease into a second disease state at a second time point; analyzing a treatment database and recommending a treatment given the first and second disease states; capturing a second liquid biopsy from the subject at a second time point; providing the second liquid biopsy to the genetic analyzer to identify the subject's genetic information; and if the genetic information from the second time point matches the predicted mutation, continuing the recommended treatment for the subject and otherwise changing the recommended treatment.
  • a method to detect abnormal cellular activities includes sequencing of cell-free nucleic acid with a genetic analyzer or a DNA sequencer; comparing current sequence reads with prior sequence reads from at least two time points; detecting a mutation of the cell-free nucleic acid and updating a diagnostic confidence indication accordingly; and detecting the presence or absence of genetic alteration and/or amount of genetic variation in an individual based on the diagnostic confidence indication of the sequence read.
  • a method for analyzing a disease state of a subject includes capturing a first liquid biopsy from the subject; providing the liquid biopsy to a genetic analyzer to identify the subject's genetic information of a first disease state at a first time point; searching for genetically similar subject profiles and predicting a mutation of the disease into a second disease state at a second time point; capturing a second liquid biopsy from the subject; providing the second liquid biopsy to a genetic analyzer to identify the subject's genetic information at a second time point; and if the genetic information from the second time point matches the predicted mutation, continuing the recommended treatment for the subject and otherwise changing the recommended treatment.
  • a method for analyzing a disease state of a subject by characterizing the subject's genetic information at two or more time points with a genetic analyzer, e.g., a DNA sequencer; and using the information from the two or more time points to produce an adjusted test result in the characterization of the subject's genetic information.
  • a genetic analyzer e.g., a DNA sequencer
  • a method detects a trend in the amount of mutation cancer polynucleotides in a sample from a subject over time by determining a frequency of the cancer polynucleotides at a plurality of time points; determining an error range for the frequency at each of the plurality of time points; determining, between an earlier and later time point, whether error ranges (1) overlap, indicating stability of frequency, (2) an increase at the later time point outside the error range, indicating increase in frequency or (3) a decrease at the later time point outside the error range, indicating decrease in frequency.
  • a method detects mutation cellular activities by sequencing of cell-free nucleic acid with a genetic analyzer, e.g., a DNA sequencer; comparing later (e.g., current) sequence reads with prior sequence reads from at least two time points and updating a diagnostic confidence indication accordingly; and detecting the presence or absence of genetic alteration and/or amount of genetic variation in an individual based on the diagnostic confidence indication of the sequence read.
  • a genetic analyzer includes any system for genetic analysis, e.g., by sequencing (DNA sequencer) or hybridization (microarray, fluorescent in situ hybridization, bionanogenomics) or other.
  • a method detects a mutation in a cell-free or substantially cell free sample obtained from a subject by generating consensus sequences by comparing later (e.g., current) sequence reads by a genetic analyzer, e.g., a DNA sequencer, with prior sequence reads from a prior period and updating a diagnostic confidence indication based on the prior sequence reads, each consensus sequence corresponding to a unique polynucleotide among a set of tagged parent polynucleotides, and generating a genetic profile of extracellular polynucleotides in the subject, wherein the genetic profile comprises a plurality of data resulting from copy number variation or mutation analyses.
  • a genetic analyzer e.g., a DNA sequencer
  • a method to detect mutation cellular activities by providing at least one set of tagged parent polynucleotides, and for each set of tagged parent polynucleotides; amplifying the tagged parent polynucleotides in the set to produce a corresponding set of amplified progeny polynucleotides; with a genetic analyzer, e.g., a DNA sequencer, sequencing a subset of the set of amplified progeny polynucleotides, to produce a set of sequencing reads; and collapsing the set of sequencing reads to generate a set of consensus sequences by comparing current sequence reads with prior sequence reads from at least one prior period and updating diagnostic confidence indication accordingly, each consensus sequence corresponding to a unique polynucleotide among the set of tagged parent polynucleotides.
  • a genetic analyzer e.g., a DNA sequencer, sequencing a subset of the set of amplified progeny polynucleotides, to
  • a method detects a mutation in a cell-free or substantially cell free sample obtained from a subject by sequencing extracellular polynucleotides from a bodily sample from a subject with a genetic analyzer, e.g., a DNA sequencer; for each of the extracellular polynucleotide, generating a plurality of sequencing reads; filtering out reads that fail to meet a set threshold; mapping sequence reads derived from the sequencing onto a reference sequence; identifying a subset of mapped sequence reads that align with a variant of the reference sequence at each mappable base position; for each mappable base position, calculating a ratio of (a) a number of mapped sequence reads that include a variant as compared to the reference sequence, to (b) a number of total sequence reads for each mappable base position; and comparing current sequence reads with prior sequence reads from at least on other time point and updating a diagnostic confidence indication accordingly.
  • a genetic analyzer e.g., a DNA sequencer
  • the method identifies one or more evolutionary paths of escape and evolved tumor treatment(s). These paths are caused by various drivers. For example, as shown in FIG. 5A , common aberrations in cancer genomes can lead to the abnormal chromosome numbers (aneuploidy) and chromosome structures of a cancer genome.
  • lines indicate the genome with germline genome on top and cancer genome with somatic aberrations below. Double lines are used when differentiating heterozygous and homozygous changes is useful. Dots represent single nucleotide changes, whereas lines and arrows represent structural changes.
  • FIG. 5B shows an exemplary system to detect the evolutionary paths of escape.
  • the system can be a Hidden Markov model (HMM), which is is a statistical Markov model in which the system being modeled is assumed to be a Markov process with unobserved (hidden) states.
  • HMM Hidden Markov model
  • an HMM can be presented as the simplest dynamic Bayesian network.
  • simpler Markov models like a Markov chain
  • the state is directly visible to the observer, and therefore the state transition probabilities are the only parameters.
  • the state In a hidden Markov model, the state is not directly visible, but output, dependent on the state, is visible.
  • Each state has a probability distribution over the possible output tokens. Therefore the sequence of tokens generated by an HMM gives some information about the sequence of states.
  • a hidden Markov model can be considered a generalization of a mixture model where the hidden variables (or latent variables), which control the mixture component to be selected for each observation, are related through a Markov process rather than independent of each other.
  • the HMM is typically defined by a set of hidden states, a matrix of state transition probabilities and a matrix of emission probabilities. Each hidden state has different statistical properties.
  • Mutations and genetic alterations including in copy number are often found to be associated with a disease state, for example, cancer. It has been observed that alterations in chromosomal copy number and loss of heterozygosity (LOH) are forms of genetic changes that often signal the activation of oncogenes and inactivation of tumor suppressor genes (anti-oncogenes). Variations in the form of copy number polymorphisms (CNP) can also occur in normal individuals. Identification of the loci implicated in these aberrations can generate anchor points which facilitate oncogenomics and toxicogenomics studies.
  • LHO chromosomal copy number and loss of heterozygosity
  • CNP copy number polymorphisms
  • the shared LOH and aberrant CN regions can be used to partition the transcriptome data and track the differential transcript expression in the affected genomic segments. Locating and exploring such alteration events is an important research approach toward understanding the cause and progression of disease.
  • the abnormal chromosomal state results when the normal diploid distribution is perturbed, resulting in changes that can include, for example, deletions, amplifications and translocations.
  • Deletions can be of a partial chromosome ranging from micro-deletions on the order of several kb to macro-deletions of mega bases, entire arms of a chromosome or entire chromosomes.
  • Amplifications can range from partial chromosomal amplifications to gains of a single copy of a chromosome to multiple copy gains of one or more chromosomes.
  • Translocations generally comprise parts of a first chromosome being translocated to another chromosome.
  • FIG. 5B shows the general architecture of an instantiated HMM for mutation detection.
  • Each oval shape X1, X2, X3 represents a random variable that can adopt any of a number of values.
  • the random variable x(t) is the hidden state at time t (x(t) ⁇ x1, x2, x3 ⁇ ).
  • the random variable y(t) is the observation at time t (with y(t) ⁇ y1, y2, y3, y4 ⁇ ).
  • the arrows in the diagram (often called a trellis diagram) denote conditional dependencies.
  • conditional probability distribution of the hidden variable x(t) at time t depends only on the value of the hidden variablex (t ⁇ 1): the values at time t ⁇ 2 and before have no influence. This is called the Markov property.
  • value of the observed variable y(t) representing the mutation conditions only depends on the value of the hidden variable x(t) (both at timet).
  • the state space of the hidden variables is discrete, while the observations themselves can either be discrete (typically generated from a categorical distribution) or continuous (typically from a Gaussian distribution).
  • the parameters of a hidden Markov model are of two types, transition probabilities and emission probabilities (also known as output probabilities).
  • the transition probabilities control the way the hidden state at time is chosen given the hidden state at time.
  • the hidden state space is assumed to consist of one of possible values, modeled as a categorical distribution. (See the section below on extensions for other possibilities.) This means that for each of the possible states that a hidden variable at time can be in, there is a transition probability from this state to each of the possible states of the hidden variable at time, for a total of transition probabilities.
  • the set of transition probabilities for transitions from any given state must sum to 1.
  • the matrix of transition probabilities is a Markov matrix. Because any one transition probability can be determined once the others are known, there are a total of transition parameters.
  • the observed variable is discrete with possible values, governed by a categorical distribution, there will be separate parameters, for a total of emission parameters over all hidden states.
  • the observed variable is an -dimensional vector distributed according to an arbitrary multivariate Gaussian distribution, there will be parameters controlling the means and parameters controlling the covariance matrix, for a total of emission parameters.
  • the HMM method can model a somatic evolution of cancer.
  • the method includes modeling genetic instability, which results in abnormal numbers of chromosomes or aneuploidy, elevated mutation rates, and altered distributions of mutational patterns.
  • the method can identify one or more cancer mutation drivers. These drivers include those that disrupt cellular signaling pathways essential for multicellular organisms and possible mutations that increase somatic fitness of cancer cells.
  • the method can include identifying dynamics of tumor progression in a population based on interactions with an environment.
  • the method includes collecting repeated genetic observations to enhance statistical inference about the evolution of tumors.
  • the method includes recommending or providing a therapeutic regimen in anticipation of the one or more escape paths.
  • Diagnosis of cancer can be done by analyzing the genetic variants, even in the presence of noise. The analysis can be based on the frequency of Sequence Variants or Level of CNV and a diagnosis confidence indication or level for detecting genetic variants in the noise range can be established. The process increases the diagnosis confidence using a plurality of measurements to increase confidence of Diagnosis (6), or alternatively using measurements at a plurality of time points to determine whether cancer is advancing, in remission or stabilized. The diagnostic confidence can be used to identify disease states.
  • cell free polynucleotides taken from a subject can include polynucleotides derived from normal cells, as well as polynucleotides derived from diseased cells, such as cancer cells.
  • Polynucleotides from cancer cells may bear genetic variants, such as somatic cell mutations and copy number variants.
  • these cancer polynucleotides are detected as sequence variants or as copy number variants.
  • the relative amount of tumor polynucleotides in a sample of cell free polynucleotides is referred to as the “tumor burden.”
  • Measurements of a parameter, whether or not they are in the noise range may be provided with a confidence interval. Tested over time, one can determine whether a cancer is advancing, stabilized or in remission by comparing confidence intervals over time. Where the confidence intervals do not overlap, this indicates the direction of disease.
  • the diagnostic confidence indication for each variant can be adjusted to indicate a confidence of predicting the observation of the CNV or mutation.
  • the confidence can be increased by using measurements at a plurality of time points to determine whether cancer is advancing, in remission or stabilized.
  • the diagnostic confidence indication can be assigned by any of a number of known statistical methods is assigned and can be based, at least in part, on the frequency at which the measurements are observed over a period of time. For example, a statistical correlation of current and prior results can be done.
  • a hidden Markov model can be built, such that a maximum likelihood or maximum a posteriori decision can be made based on the frequency of occurrence of a particular test event from a plurality of measurements or a time points.
  • the probability of error and resultant diagnostic confidence indication for a particular decision can be output as well.
  • the measurements of a parameter, whether or not they are in the noise range may be provided with a confidence interval. Tested over time, one can increase the predictive confidence of whether a cancer is advancing, stabilized or in remission by comparing confidence intervals over time. Two time points can be separated by about a month to about a year, about a year to about 5 years, or no more than about three months.
  • the HMM detect with high sensitivity genetic variation in a sample of initial genetic material.
  • the methods involve using one to three of the following tools: First, the efficient conversion of individual polynucleotides in a sample of initial genetic material into sequence-ready tagged parent polynucleotides, so as to increase the probability that individual polynucleotides in a sample of initial genetic material will be represented in a sequence-ready sample. This can produce sequence information about more polynucleotides in the initial sample.
  • Second, high yield generation of consensus sequences for tagged parent polynucleotides by high rate sampling of progeny polynucleotides amplified from the tagged parent polynucleotides, and collapsing of generated sequence reads into consensus sequences representing sequences of parent tagged polynucleotides. This can reduce noise introduced by amplification bias and/or sequencing errors, and can increase sensitivity of detection. Third, the noise in the detection of mutations and copy number variations is reduced by comparing prior sample analysis with the current sample and increasing a diagnostic confidence indication if the same mutations and copy number variations have appeared in prior analysis and otherwise decreasing the diagnostic confidence indication if this is the first time the sequence is observed.
  • FIG. 5C shows an exemplary model generated by the system of FIG. 2B for inferring tumor phylogeny from next-generation sequencing data.
  • the subclones are related to each other by an evolutionary process of acquisition of mutations.
  • the three clones (leaf nodes) are characterized by different combinations of the four single nucleotide variant (SNV) sets A, B, C, and D.
  • SNV single nucleotide variant
  • the percentages on the edges of the tree indicate the fraction of cells with this particular set of SNVs, e.g., 70% of all cells carry A, 40% additionally carry B, and only 7% carry A, B, and D.
  • FIG. 5D shows an exemplary a heterogeneous collection of normal cells and cancer subclones developed during an evolutionary history of a tumor.
  • the evolutionary history of a tumor gives rise to a heterogeneous collection of normal cells (small discs) and cancer subclones (large discs, triangles, squares). Internal nodes that have been fully replaced by their descendants (like the one carrying SNV sets A and B without C or D) are no longer part of the tumor.
  • Embodiments of the invention can take the form of an entirely hardware embodiment, an entirely software embodiment or an embodiment containing both hardware and software elements.
  • the invention is implemented in software, which includes but is not limited to firmware, resident software, microcode, and the like.
  • the invention can take the form of a computer program product accessible from a computer-usable or computer-readable medium providing program code for use by or in connection with a computer or any instruction execution system.
  • a computer-usable or computer readable medium can be any apparatus that can contain, store, communicate, or transport the program for use by or in connection with the instruction execution system, apparatus, or device.
  • the medium can be an electronic, magnetic, optical, electromagnetic, infrared, or semiconductor system (or apparatus or device).
  • Examples of a computer-readable medium include a semiconductor or solid state memory, magnetic tape, a removable computer diskette, a random access memory (RAM), a read-only memory (ROM), a rigid magnetic disk and an optical disk.
  • Current examples of optical disks include compact disk-read only memory (CD-ROM), compact disk-read/write (CD-R/W) and DVD.
  • a data processing system suitable for storing and/or executing program code will include at least one processor coupled directly or indirectly to memory elements through a system bus.
  • the memory elements can include local memory employed during actual execution of the program code, bulk storage, and cache memories which provide temporary storage of at least some program code in order to reduce the number of times code must be retrieved from bulk storage during execution.
  • I/O devices including but not limited to keyboards, displays, pointing devices, etc.
  • Network adapters may also be coupled to the system to enable the data processing system to become coupled to other data processing systems or remote printers or storage devices through intervening private or public networks. Modems, cable modem and Ethernet cards are just a few of the currently available types of network adapters.

Abstract

A system is disclosed for personalized medical treatment based on biomarkers.

Description

    BACKGROUND
  • The present invention relates to the field of computerized drug identification.
  • Drug interactions remain a principal aspect of the pharmaceutical sciences. A drug interaction is a commonly known situation in which a substance affects the activity of a drug, such that the effects of a given drug is increased or decreased, or the combination of the substance and the drug produce a new effect that neither produces alone. Typically, drug-drug interactions are most unpredictable; however, drug-food interactions also are known to exist between drugs and foods, as well as drug-herb interactions between drugs and herbs.
  • Generally speaking, it is desirable to avoid drug interactions due to the possibility of a poor or unexpected outcome resulting from the interaction of a drug with another substance. Consequently, known drug interactions often are listed in the literature distributed with a drug. Providing an exhaustive list of drug interactions in literature, however, can be difficult when a substantial number of drug interactions are known to exist. As such, voluminous books have been created as an aggregation of known drug interactions. While the most diligent review of a book of known drug interactions will reveal the requisite information necessary to avoid an undesirable outcome from a drug interaction of a prescribed selection of drugs, in practice it is not reasonable to presume that a dispensary of drugs will consult the requisite literature when dispensing a drug.
  • SUMMARY
  • In one aspect, a system includes code to match genomic biomarker(s) from gene or DNA sequencing for a population with historical information for a population on drug structure, dosage, clinical variability and risk for adverse events for the drug substance, the computer constructing side effect features for each drug, and a classifier to predict whether a drug pair results in adverse interactions, the computer generating one or more indicia for the drug substance; and code to apply the indicia to select a custom dosage for a drug whose properties are selected and crafted for the individual patient to provide personalized medicine.
  • In another aspect, a system includes code to: acquire subject genetic scans using a gene sequencer; identify each substance to be provided to the subject; determine substance interactions for each drug and the genetic scan; and provide indicia associated with each substance to warn the subject or a medical provider based on the genetic scan.
  • In another aspect, a system includes a substance to be consumed by a subject and one or more indicia labeling the substance with: genomic biomarkers; drug exposure and clinical response variability; risk for adverse events; genotype-specific dosing; polymorphic drug target and disposition genes; and treatment based on the biomarker.
  • Advantages of the system may include one or more of the following. The system may make medical trials more efficient. This will lower the costs that come about due to adverse drug side effects and prescription of drugs that have been proven ineffective in certain genotypes. Drug companies can develop and license a drug specifically intended for those who are the small population genetically at risk for adverse side effects.
  • Additional aspects of the invention will be set forth in part in the description which follows, and in part will be obvious from the description, or may be learned by practice of the invention. The aspects of the invention will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The accompanying drawings, which are incorporated in and constitute part of this specification, illustrate embodiments of the invention and together with the description, serve to explain the principles of the invention. The embodiments illustrated herein are presently preferred, it being understood, however, that the invention is not limited to the precise arrangements and instrumentalities shown, wherein:
  • FIGS. 1A-1B show exemplary substance interaction management processes;
  • FIG. 2 is a schematic illustration of a data processing system configured for computer management genetic information, precision medication and drug interaction information retrieval; and,
  • FIG. 3 is a flow chart illustrating a process for pharmacogenetics drug interaction information retrieval.
  • FIG. 4 shows a big data learning machine to process genetic data and determine pharmacogenetics relationship among genes and drugs for drug interaction purposes.
  • FIG. 5A shows various common aberrations in cancer genomes.
  • FIG. 5B shows an exemplary system to detect the evolutionary paths of escape.
  • FIG. 5C shows an exemplary model generated by the system of FIG. 5B.
  • FIG. 5D shows an exemplary a heterogeneous collection of normal cells and cancer subclones developed during an evolutionary history of a tumor.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Embodiments of the present invention provide a method, system and computer program product for computer identification (scanning or imaging of drugs) for drug interaction information retrieval. In accordance with an embodiment of the present invention, multiple different drugs can be scanned or imaged to detect identifiable content disposed on the different drugs. Each drug can be compared to a data store of drug information to identify each drug. Thereafter, pharmacogenetics data and drug interaction data can be retrieved for each identified drug. Further, known drug-drug interactions and genetic impacts for the identified drugs can be determined and a report can be provided to include the known drug-drug interactions. In this way, precision medicine and drug-drug interactions resulting from the use of the multiple different drugs can be determined without recourse to a voluminous text of drug interactions.
  • In illustration, FIGS. 1A-1B show exemplary processes for computer detection of substances or drugs for drug interaction information retrieval. As shown in FIG. 1A, in 10, the process cryptographically reads substance content from RF tag or barcode on a secure bottle and in 20 identifies content for the substance. In 30, identified substances are added to an interaction list and in 40 the process determines if additional substances remain to be scanned (RF/Bar Code), and if so loops back to 10. In 50, the interaction list now populated by a list of scanned substances is processed. Next, in 60 the process retrieves drug data and drug interaction data for each of the scanned substances and determines relative interactions between substances. In 70, the process receives genetic scans for subjects. In 80, the process applies pharmacogenomic information to the drug-gene interaction data and selects the best medication and identify people who need an unusually high or low dose. In 90, relative interactions can be rendered within a report such as a paper report or a graphical user interface display.
  • Turning now to FIG. 1B, a camera based drug identification and drug interaction management system is shown. In 110, images of a substance such as a drug can be acquired. In 120 the process identifies content for the substance can be retrieved from the image. In 130, substances can be identified according to the identifying content. In 140, the identified substance can be added to an interaction list. In 150 the process loops back to 110 if additional substances remain to be imaged and if not, the interaction list now populated by a list of imaged substances can be processed. In 160, the process receives genetic scans for subjects. In 170, the process retrieves drug data and drug interaction data for each of the imaged substances and determines relative interactions between substances, and in 180 pharmacogenomic information is applied to select the best medication and identify people who need an unusually high or low dose. In 190, the relative interactions can be rendered within a report such as a paper report or a graphical user interface display.
  • The process shown in FIGS. 1A-1B can be implemented within a data processing system. In further illustration, FIG. 2 schematically depicts a data processing system configured for computer visualization of drugs for drug interaction information retrieval. The system can include a host computing platform 202 coupled to a camera 220 such as a digital still camera or digital video camera. The camera 220 can be focused on a marshalling point 240 provided by a marshalling apparatus 230, for example gravity feed or isolation chamber or miniature conveyor belt. The host computing platform 202 also can be communicatively coupled a drug image data store 250 of known substances and corresponding known identifying content visually disposed on the known substances. The host computing platform 202 additionally can be communicatively coupled to a drug interaction data store 260 providing drug interaction data for different substances relative to other substances including prescription and over-the-counter drugs, vitamins and herbal remedies, and food products.
  • In one embodiment of FIG. 2, multiple different substances such as prescription drugs, over-the-counter drugs or even vitamins and herbal remedies can be provided to a marshalling apparatus such as a gravity feed or miniature conveyor belt or even a chamber. The marshalling apparatus can isolate an individual one of the different substances for imaging by camera 220, for example a charge coupled device (CCD) driven digital camera or video recorder. The camera 220 can capture an image of each individual one of the different substances 110A, 110B, 110N and computer visualization for drug interaction information retrieval logic 300 can process each captured image to detect identifying content disposed on each of the different substances such as a pill marking or code. The computer visualization for drug interaction information retrieval logic 300 in turn can compare the identified content to a data store of known substances 140 to identify each of the different substances. The computer can lookup not only known drug interactions for each of the different substances, but also known drug interactions between the identified ones of the substances and pharmacogenetics impact on the individual patient. Thereafter, a drug interaction report can be produced indicating the known drug interactions between the identified ones of the substances.
  • The host computing platform 202 can support the execution of computer scanning or visualization for drug interaction information retrieval logic 270. The logic can include program code enabled to acquire imagery of different substances in the marshalling point 240. The program code further can be enabled to locate and retrieve identifying content disposed on the different substances and to look up the identifying content in the drug image data store 250 in order to identify each of the substances. The program code yet further can be enabled to retrieve from drug interaction data store 260 drug interactions for each of the identified substances and to particularly correlate the retrieved drug interactions to different ones of the substances so that relative drug interactions can be determined for the substances. Finally, the program code can be enabled to render a report of drug interaction data in a graphical user interface display 280 of drug interaction data.
  • The computing platform 202 also receives pharmacogenetics interaction 282. Notably, the host computing platform 202 can support the execution of computer visualization for pharmacogenetics interaction information retrieval logic 272. Genetic information is captured by high speed gene sequencing machine 210 that uploads gene data to a cloud computing network 212. The doctors, pharmacists, or consumers can access DNA information using mobile computers such as smart phone 214, for example.
  • The system can have wireless communication 292 with the medication's labels. For example, the labels can have RF tags or NFC tags that provide upon inquiry FDA required labeling contents. In one embodiment, the content can be genomic biomarkers; drug exposure and clinical response variability; risk for adverse events; genotype-specific dosing; polymorphic drug target and disposition genes; and treatment based on the biomarker. NFC tags are passive devices and operate without a power supply of their own and are reliant on an active device to come into range before they are activated. To power these NFC tags, electromagnetic induction is used to create a current in the passive device. Active devices, such as a reader or a smartphone, are responsible for generating the magnetic field with a simple coil of wire, which produces magnetic fields perpendicular to the flow of the alternating current in the wire. To reduce power, NFC operates over just a few inches, rather than the meters in other types of wireless communication.
  • The system can be used to provide personalized medicine through custom pharmacy compounding 294 or custom production of a drug whose various properties (e.g. dose level, ingredient selection, route of administration, etc.) are selected and crafted for an individual patient (in contrast to mass-produced unit doses or fixed-dose combinations).
  • The genetic scan in 70 can be generated by gene sequencing machines. DNA sequencing is the process of determining the precise order of nucleotides within a DNA molecule. It includes any method or technology that is used to determine the order of the four bases—adenine, guanine, cytosine, and thymine—in a strand of DNA. Various high speed sequencers can be used. For example, Nanopore DNA sequencing is based on the readout of electrical signals occurring at nucleotides passing by alpha-hemolysin pores covalently bound with cyclodextrin. The DNA passing through the nanopore changes its ion current. Oxford Nanopore Technologies offers a handheld sequencer capable of generating more than 150 megabases of sequencing data in one run.
  • Another approach uses measurements of the electrical tunnelling currents across single-strand DNA as it moves through a channel. Depending on its electronic structure, each base affects the tunnelling current differently, allowing differentiation between different bases. The use of tunnelling currents has the potential to sequence orders of magnitude faster than ionic current methods and the sequencing of several DNA oligomers and micro-RNA has already been achieved. Sequencing by hybridization is a non-enzymatic method that uses a DNA microarray. A single pool of DNA whose sequence is to be determined is fluorescently labeled and hybridized to an array containing known sequences. Strong hybridization signals from a given spot on the array identify its sequence in the DNA being sequenced. Mass spectrometry may be used to determine DNA sequences. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry, or MALDI-TOF MS, has specifically been investigated as an alternative method to gel electrophoresis for visualizing DNA fragments. With this method, DNA fragments generated by chain-termination sequencing reactions are compared by mass rather than by size. The mass of each nucleotide is different from the others and this difference is detectable by mass spectrometry. Single-nucleotide mutations in a fragment can be more easily detected with MS than by gel electrophoresis alone. MALDI-TOF MS can more easily detect differences between RNA fragments, so researchers may indirectly sequence DNA with MS-based methods by converting it to RNA first. In microfluidic Sanger sequencing the entire thermocycling amplification of DNA fragments as well as their separation by electrophoresis is done on a single glass wafer (approximately 10 cm in diameter) thus reducing the reagent usage as well as cost. Microscopy-based technique directly visualizes the sequence of DNA molecules using electron microscopy. RNAP sequencing is based on use of RNA polymerase (RNAP), which is attached to a polystyrene bead. One end of DNA to be sequenced is attached to another bead, with both beads being placed in optical traps. RNAP motion during transcription brings the beads in closer and their relative distance changes, which can then be recorded at a single nucleotide resolution. The sequence is deduced based on the four readouts with lowered concentrations of each of the four nucleotide types, similarly to the Sanger method. Other high speed gene sequencers can be used.
  • The system applies pharmacogenomic information to select the best medication and identify people who need an unusually high or low dose. This is in addition to clinical factors, such as a patient's age, weight, sex, and liver and kidney function. Pharmacogenomics (sometimes called pharmacogenetics) is focused on understanding how genes affect individual responses to medications and to help doctors select the drugs and dosages best suited for each person. Pharmacogenomics looks at variations in genes for proteins that influence drug responses. Such proteins include a number of liver enzymes that convert medications into their active or inactive forms. Even small differences in the genetic sequences of these enzymes can have a big impact on a drug's safety or effectiveness. One example involves a liver enzyme known as CYP2D6. This enzyme acts on a quarter of all prescription drugs, including the painkiller codeine, which it converts into the drug's active form, morphine. The CYP2D6 gene exists in more than 160 different versions, many of which vary by only a single difference in their DNA sequence, although some have larger changes. The majority of these variants don't affect drug responses. Some people have hundreds or even thousands of copies of the CYP2D6 gene (typically, people have two copies of each gene). Those with extra copies of this gene manufacture an overabundance of CYP2D6 enzyme molecules and metabolize the drug very rapidly. As a result, codeine may be converted to morphine so quickly and completely that a standard dose of the drug can be an overdose. On the other end of the spectrum, some variants of CYP2D6 result in a nonfunctional enzyme. People with these variants metabolize codeine slowly, if at all, so they might not experience much pain relief. For these people, doctors might prescribe a different type of pain reliever. Pharmacogenomic information can cover dosage guidance, possible side effects or differences in effectiveness for people with certain genomic variations—can help doctors tailor their drug prescriptions for individual patients. The system applies pharmacogenomic data to develop and market drugs for people with specific genetic profiles. The system can identify the genetic basis for certain serious side effects, drugs could be prescribed only to people who are not at risk for them. As a result, potentially lifesaving medications, which otherwise might be taken off the market because they pose a risk for some people, could still be available to those who could benefit from them. For example, a few drug and gene associations are listed in the Appendix.
  • It will be recognized by the skilled artisan that while the computer visualization for drug interaction information retrieval logic 270 is shown to execute in a single host computing platform 202, the invention is not so limited and the computer visualization for drug interaction information retrieval logic 270 also can be distributed in form across multiple different computing platforms. Further, the camera 220 and marshalling apparatus 230 can be located remotely from the host computing platform 202 whilst providing acquired imagery to the host computing platform 210 over a computer communications network, whether wireless or wirebound. Yet further, either or both of the drug image data store 250 and the drug interaction data store 260 can be remotely disposed from the host computing platform 202 and accessible over a computer communications network, whether wireless or wirebound.
  • The system enables a medical model that separates patients into different groups—with medical decisions, practices, interventions and/or products being tailored to the individual patient based on their predicted response or risk of disease.
  • Having the ability to look at a patient on an individual basis will allow for a more accurate diagnosis and specific treatment plan. Genotyping is the process of obtaining an individual's DNA sequence by using biological assays. By having a detailed account of an individual's DNA sequence, their genome can then be compared to a reference genome, like that of the Human Genome Project, to assess the existing genetic variations that can account for possible diseases. An individual's genetic make-up also plays a large role in how well they respond to a certain treatment, and therefore, knowing their genetic content can change the type of treatment they receive. The system applies pharmacogenomics by using an individual's genome to provide a more informed and tailored drug prescription. Often, drugs are prescribed with the idea that it will work relatively the same for everyone, but in the application of drugs, there are a number of factors that must be considered. The detailed account of genetic information from the individual will help prevent adverse events, allow for appropriate dosages, and create maximum efficacy with drug prescriptions. The pharmacogenomic process for discovery of genetic variants that predict adverse events to a specific drug has been termed toxgnostics.
  • In addition to specific treatment, personalized medicine can greatly aid the advancements of preventive care. For instance, many women are already being genotyped for certain mutations in the BRCA1 and BRCA2 gene if they are predisposed because of a family history of breast cancer or ovarian cancer. As more causes of diseases are mapped out according to mutations that exist within a genome, the easier they can be identified in an individual. Measures can then be taken to prevent a disease from developing. Even if mutations were found within a genome, having the details of their DNA can reduce the impact or delay the onset of certain diseases. Having the genetic content of an individual will allow better guided decisions in determining the source of the disease and thus treating it or preventing its progression. This will be extremely useful for diseases like Alzheimer's or cancers that are thought to be linked to certain mutations in human DNA.
  • The system can be used to test efficacy and safety of a drug specific to a targeted patient group/sub-group is companion diagnostics. This technology is an assay that is developed during or after a drug is made available on the market and is helpful in enhancing the therapeutic treatment available based on the individual. These companion diagnostics have incorporated the pharmacogenomic information related to the drug into their prescription label in an effort to assist in making the most optimal treatment decision possible for the patient.
  • Having an individual's genomic information can be significant in the process of developing drugs as they await approval from the FDA for public use. Having a detailed account of an individual's genetic make-up can be a major asset in deciding if a patient can be chosen for inclusion or exclusion in the final stages of a clinical trial. Being able to identify patients who will benefit most from a clinical trial will increase the safety of patients from adverse outcomes caused by the product in testing, and will allow smaller and faster trials that lead to lower overall costs. In addition, drugs that are deemed ineffective for the larger population can gain approval by the FDA by using personal genomes to qualify the effectiveness and need for that specific drug or therapy even though it may only be needed by a small percentage of the population. Treatments can be more specifically tailored to an individual and give insight into how their body will respond to the drug and if that drug will work based on their genome. The personal genotype can allow physicians to have more detailed information that will guide them in their decision in treatment prescriptions, which will be more cost-effective and accurate.
  • FIG. 3 shows a method 300 for predicting drug-drug interactions based on genetic data and clinical side effects, in accordance with an embodiment of the present principles. The process includes the following: At 310, construct a comprehensive gene-drug-drug interactions (GDDIs) training dataset that includes all pharmaceutical, pharmacokinetic (PK), pharmacogenetic (PG), and pharmacodynamic (PD) GDDIs from multiple data sources for each drug in a set of drugs under consideration. In an embodiment, the multiple data sources can include, but are not limited to, the following: gene sequencers, clinical trials; drug development information; empirical information; a drug bank; drug label information; an adverse event reporting system (e.g., the FDA Adverse Event Reporting System information (FAERS)); and text mining from scientific documents (e.g., search tool for interactions of chemicals (STITCH)). At step 320, construct side effect features for each of the drugs in the set from genetic panels for an individual and side effects associated with the drugs in the set. In an embodiment, the genetic panels are generated by genetic sequencers, and all drugs' side effects, from which the side effect features are constructed, come from one or more of the following sources: clinical trials; drug development; empirical information; FDA drug label (SIDER and DAILYMED®); FDA Adverse Event Reporting System (FAERS); and real-world evidence. At 330, build, using the GDDIs training dataset, a GDDIs classifier for predicting whether or not a given drug pair derived from the set of drugs results in adverse interactions, and repeat this process for all possible drug pairs derivable from the set of drugs. In an embodiment, the features used for building the classifier can include, but are not limited to, the following: drug's clinical side effect keywords; and other drug properties (e.g., chemical structures, protein targets, and so forth).
  • At 340, obtain predicted GDDIs from the classifier. At 350, for each side effect, perform statistical test to determine whether that side effect is differentially shown between positive predicted GDDIs and negative predicted GDDIs. In one embodiment, the term “positive predicted GDDIs” refers to drugs pairs that cannot be taken together given a patient genetic profile. In contrast, the term “negative predicted GDDIs” refers to drugs pairs that may be safe to use together with a genetic profile.
  • Side effects are effects after taking a medicine, which are other than the intended therapeutic effects. Label side effects means the side effects are recorded in drug labels (for example, but not limited to, SIDER database, DAILYMED®, and so forth). FDA side effects means the side effects are recorded in, for example, but not limited to, the FDA Adverse Event Reporting System (FAERS). Consider, for example, the drug Ibuprofen as an example, DAILYMED® records its 249 types of label side effects (e.g., abdominal discomfort, confusion, dry mouth, vomiting, and weight loss), and FAERS records its 728 types of FDA side effects (e.g., anxiety, ear ache, fatigue, tooth loss, sleep disorder).
  • In 380, relative interactions between the different drug substances can be determined by locating references in the interaction data for each of the drug substances to others of the substances. Finally, in block 390, the relative interactions can be rendered within a report such as a paper report or a graphical user interface display. Optionally, an activatable link can be provided in the display for selected ones of the drug substances for reordering the selected ones of the drug substances. In this way, the relative drug interactions resulting from the dispensing of multiple different drug substances based on patient genetic data can be determined without requiring a tedious manual process of looking up drug interaction data for each substance and manually correlating the drug interaction data for the specific combination of dispensed substances.
  • The system can also perform GDDI discovery and prediction that uses molecular structure similarity information derived from fingerprint-based modeling. Identifying new GDDIs using structural similarity is based on the basic idea that if drug A interacts with drug B, and drug C is structurally similar to A, then C should also interact with B (the argument also follows if A is replaced with B). Hence, by combining knowledge of known interactions with structural similarity it is possible to identify new interactions. The process uses a list of drug-drug interactions from DrugBank (step 1), structural similarity computation was carried out using molecular fingerprints (step 2), apply gene-drug interaction to similar drugs, and a new list of gene-drug interactions can be inferred.
  • Structural similarity can be identified in three steps: 1) Collecting and processing drug structures: Information on the structures of the compounds in DrugBank is retrieved along with the SMILE code (a chemical notation representing a chemical structure in linear textual form). 2) Structural representation: BIT_MACCS (MACCS Structural Keys Bit packed) fingerprints are calculated for all molecules included in the study and each molecule is represented as a bit vector that codes the presence or absence of structural features where each feature is assigned a specific bit position. 3) Similarity measures, computation, and data representation: Different measures are used to compare similarity between two molecular fingerprints. In one embodiment, the molecular fingerprints were compared using Tanimoto coefficient (TC). The TC can span values between 0 and 1, where 0 means ‘maximum dissimilarity’ and 1 means ‘maximum similarity.’ The TC between two fingerprint representations A and B is defined as the number of features present in the intersection of both fingerprints A and B divided by the number of features present in the union of both fingerprints. Next, for each drug affected by a particular gene, the process predicts new gene based DDIs. One embodiment predicts new DDIs reduces to matrix multiplication of the matrices M1, which consists of the established interactions, and M2, which consists of the similarity matrix.
  • The pharmacogenomic information can be applied to drug labeling. One embodiment may contain information on genomic biomarkers and can describe:
      • Drug exposure and clinical response variability
      • Risk for adverse events
      • Genotype-specific dosing
      • Mechanisms of drug action
      • Polymorphic drug target and disposition genes
  • The information may include specific actions to be taken based on the biomarker information. Pharmacogenomic information can appear in different sections of the labeling depending on the actions. Biomarkers in the table include but are not limited to germ-line or somatic gene variants, functional deficiencies, expression changes, and chromosomal abnormalities; selected protein biomarkers that are used to select patients for treatment are also included.
  • In one embodiment, the process includes constructing a gene-drug interactions training dataset that includes pharmaceutical, pharmacokinetic or pharmacodynamics, and pharmacogenomics drug-drug interactions for each drug; constructing side effect features for each of the plurality of drugs from side effects associated with the plurality of drugs; running a gene-drug-drug interactions classifier that predicts adverse drug-drug interactions for drug pairs and the genetic scan; and for each of the side effects, performing a Fisher's exact test to determine predicted gene-drug-drug interactions. Fisher's exact testis a statistical significance test used in the analysis of contingency tables. It is one of a class of exact tests, so called because the significance of the deviation from a null hypothesis (e.g., P-value) can be calculated exactly, rather than relying on an approximation that becomes exact in the limit as the sample size grows to infinity, as with many statistical tests.
  • FIG. 4 shows a deep learning machine using deep convolutionary neural networks for detecting genetic based drug-drug interaction. One embodiment uses an AlexNet: 8-layer architecture, while another embodiment uses a VGGNet: 16-layer architecture (each pooling layer and last 2 FC layers are applied as feature vector). For drugs, the indications of use and other drugs used capture most of many important covariates. One embodiment access data from SIDER (a text-mined database of drug package inserts), the Offsides database that contains information complementary to that found in SIDER and improves the prediction of protein targets and drug indications, and the Twosides database of mined putative DDIs also lists predicted adverse events, all available at the http://PharmGKB.org Web site.
  • The system of FIG. 4 receives data on adverse events strongly associated with indications for which the indication and the adverse event have a known causative relationship. A drug-event association is synthetic if it has a tight reporting correlation with the indication (ρ≥0.1) and a high relative reporting (RR) association score (RR≥2). Drugs reported frequently with these indications were 80.0 (95% CI, 14.2 to 3132.8; P<0.0001, Fisher's exact test) times as likely to have synthetic associations with indication events. Disease indications are a significant source of synthetic associations. The more disproportionately a drug is reported with an indication (x axis), the more likely that drug will be synthetically associated. For example, adverse events strongly associated with drugs are retrieved from the drug's package insert. These drug-event pairs represent a set of known strong positive associations.
  • Adverse events related to sex and race are also analyzed. For example, for physiological reasons, certain events predominantly occur in males (for example, penile swelling and azoospermia). Drugs that are disproportionately reported as causing adverse events in males were more likely to be synthetically associated with these events. Similarly, adverse events that predominantly occur in either relatively young or relatively old patients are analyzed.
  • “Off-label” adverse event data is also analyzed, and off-label uses refer to any drug effect not already listed on the drug's package insert. For example, the SIDER database, extracted from drug package inserts, lists 48,577 drug-event associations for 620 drugs and 1092 adverse events that are also covered by the data mining. Offsides recovers 38.8% (18,842 drug-event associations) of SIDER associations from the adverse event reports. Thus, Offsides finds different associations from those reported during clinical trials before drug approval.
  • Polypharmacy side effects for pairs of drugs (Twosides) are also analyzed. These associations are limited to only those that cannot be clearly attributed to either drug alone (that is, those associations covered in Offsides). The database contains an significant associations for which the drug pair has a higher side-effect association score, determined using the proportional reporting ratio (PRR), than those of the individual drugs alone. The system determines pairwise similarity metrics between all drugs in the Offsides and SIDER databases. The system can predict shared protein targets using drug-effect similarities. The side-effect similarity score between two drugs is linearly related to the number of targets that those drugs share.
  • The system can determine relationships between the proportion of shared indications between a pair of drugs and the similarity of their side-effect profiles in Offsides. The system can use side-effect profiles to suggest new uses for old drugs. While the preferred system predicts existing therapeutic indications of known drugs, the system can recommend drug repurposing using drug-effect similarities in Offsides.
  • Corroboration of class-wide interaction effects with EMRs. The system can identify DDIs shared by an entire drug class. The class-class interaction analysis generates putative drug class interactions. The system analyzes laboratory reports commonly recorded in EMRs that may be used as markers of these class-specific DDIs.
  • The system can be used systematic drug surveillance. The FDA manages a collection of adverse drug event reports to monitor the safety of drugs. They rely on physicians, pharmaceutical companies, and patients to volunteer these reports. Since reporting is not mandatory, many adverse drug events that occur are never reported to the FDA. To address this issue, an embodiment of the present invention uses an algorithm to infer unreported adverse drug events. This embodiment relies on the fact that many adverse events occur together. For example, nausea and vomiting commonly manifest together. Therefore, if a drug is observed to causes nausea, it can be inferred that it also causes vomiting.
  • The successful prediction of side effects before a drug enters clinical trials can be done. Chemical informatics techniques can predict drug side effects by comparing the structural similarity of drugs. Protein structural similarity is learned by the deep learning system to predict drug side effects. More recently, network and chemical properties are used for predictive models of drug effects and leverage the system's comprehensive database of known drug effects.
  • In a parallel trend, anti-biotics and cancer treatments have lost their effect over time. As such, even though there is no adverse event, there is still a negative consequence for the patient when the virus/tumor develops resistance to the drug. Thus, the processor can analyze evolutions in the target of the treatment and recommend alternative treatment.
  • In one aspect, systems and methods includes analyzing a disease state of a subject by collecting genetic profile data on a population of tumors and original tumor treatment(s); identifying one or more evolutionary paths of escape and evolved tumor treatment(s); and based on a subject profile, predicting a probability of escape along the one or more evolutionary paths.
  • In another aspect, a method for analyzing a disease state of a subject includes capturing a first liquid biopsy from the subject; providing the liquid biopsy to a genetic analyzer to identify the subject's genetic information of a first disease state at a first time point; searching for genetically similar patients and predicting a mutation of the disease into a second disease state at a second time point; analyzing a treatment database and recommending a treatment given the first and second disease states; capturing a second liquid biopsy from the subject at a second time point; providing the second liquid biopsy to the genetic analyzer to identify the subject's genetic information; and if the genetic information from the second time point matches the predicted mutation, continuing the recommended treatment for the subject and otherwise changing the recommended treatment.
  • In yet another aspect, a method to detect abnormal cellular activities includes sequencing of cell-free nucleic acid with a genetic analyzer or a DNA sequencer; comparing current sequence reads with prior sequence reads from at least two time points; detecting a mutation of the cell-free nucleic acid and updating a diagnostic confidence indication accordingly; and detecting the presence or absence of genetic alteration and/or amount of genetic variation in an individual based on the diagnostic confidence indication of the sequence read.
  • In a further aspect, a method for analyzing a disease state of a subject includes capturing a first liquid biopsy from the subject; providing the liquid biopsy to a genetic analyzer to identify the subject's genetic information of a first disease state at a first time point; searching for genetically similar subject profiles and predicting a mutation of the disease into a second disease state at a second time point; capturing a second liquid biopsy from the subject; providing the second liquid biopsy to a genetic analyzer to identify the subject's genetic information at a second time point; and if the genetic information from the second time point matches the predicted mutation, continuing the recommended treatment for the subject and otherwise changing the recommended treatment.
  • In another aspect disclosed herein is a method for analyzing a disease state of a subject by characterizing the subject's genetic information at two or more time points with a genetic analyzer, e.g., a DNA sequencer; and using the information from the two or more time points to produce an adjusted test result in the characterization of the subject's genetic information.
  • In another aspect, a method detects a trend in the amount of mutation cancer polynucleotides in a sample from a subject over time by determining a frequency of the cancer polynucleotides at a plurality of time points; determining an error range for the frequency at each of the plurality of time points; determining, between an earlier and later time point, whether error ranges (1) overlap, indicating stability of frequency, (2) an increase at the later time point outside the error range, indicating increase in frequency or (3) a decrease at the later time point outside the error range, indicating decrease in frequency.
  • In yet another aspect, a method detects mutation cellular activities by sequencing of cell-free nucleic acid with a genetic analyzer, e.g., a DNA sequencer; comparing later (e.g., current) sequence reads with prior sequence reads from at least two time points and updating a diagnostic confidence indication accordingly; and detecting the presence or absence of genetic alteration and/or amount of genetic variation in an individual based on the diagnostic confidence indication of the sequence read. A genetic analyzer includes any system for genetic analysis, e.g., by sequencing (DNA sequencer) or hybridization (microarray, fluorescent in situ hybridization, bionanogenomics) or other.
  • In another aspect, a method detects a mutation in a cell-free or substantially cell free sample obtained from a subject by generating consensus sequences by comparing later (e.g., current) sequence reads by a genetic analyzer, e.g., a DNA sequencer, with prior sequence reads from a prior period and updating a diagnostic confidence indication based on the prior sequence reads, each consensus sequence corresponding to a unique polynucleotide among a set of tagged parent polynucleotides, and generating a genetic profile of extracellular polynucleotides in the subject, wherein the genetic profile comprises a plurality of data resulting from copy number variation or mutation analyses.
  • In another aspect disclosed herein is a method to detect mutation cellular activities by providing at least one set of tagged parent polynucleotides, and for each set of tagged parent polynucleotides; amplifying the tagged parent polynucleotides in the set to produce a corresponding set of amplified progeny polynucleotides; with a genetic analyzer, e.g., a DNA sequencer, sequencing a subset of the set of amplified progeny polynucleotides, to produce a set of sequencing reads; and collapsing the set of sequencing reads to generate a set of consensus sequences by comparing current sequence reads with prior sequence reads from at least one prior period and updating diagnostic confidence indication accordingly, each consensus sequence corresponding to a unique polynucleotide among the set of tagged parent polynucleotides.
  • In yet another aspect, a method detects a mutation in a cell-free or substantially cell free sample obtained from a subject by sequencing extracellular polynucleotides from a bodily sample from a subject with a genetic analyzer, e.g., a DNA sequencer; for each of the extracellular polynucleotide, generating a plurality of sequencing reads; filtering out reads that fail to meet a set threshold; mapping sequence reads derived from the sequencing onto a reference sequence; identifying a subset of mapped sequence reads that align with a variant of the reference sequence at each mappable base position; for each mappable base position, calculating a ratio of (a) a number of mapped sequence reads that include a variant as compared to the reference sequence, to (b) a number of total sequence reads for each mappable base position; and comparing current sequence reads with prior sequence reads from at least on other time point and updating a diagnostic confidence indication accordingly.
  • The method identifies one or more evolutionary paths of escape and evolved tumor treatment(s). These paths are caused by various drivers. For example, as shown in FIG. 5A, common aberrations in cancer genomes can lead to the abnormal chromosome numbers (aneuploidy) and chromosome structures of a cancer genome. In FIG. 5A, lines indicate the genome with germline genome on top and cancer genome with somatic aberrations below. Double lines are used when differentiating heterozygous and homozygous changes is useful. Dots represent single nucleotide changes, whereas lines and arrows represent structural changes.
  • FIG. 5B shows an exemplary system to detect the evolutionary paths of escape. The system can be a Hidden Markov model (HMM), which is is a statistical Markov model in which the system being modeled is assumed to be a Markov process with unobserved (hidden) states. As known to those skilled in the art, an HMM can be presented as the simplest dynamic Bayesian network. In simpler Markov models (like a Markov chain), the state is directly visible to the observer, and therefore the state transition probabilities are the only parameters. In a hidden Markov model, the state is not directly visible, but output, dependent on the state, is visible. Each state has a probability distribution over the possible output tokens. Therefore the sequence of tokens generated by an HMM gives some information about the sequence of states. A hidden Markov model can be considered a generalization of a mixture model where the hidden variables (or latent variables), which control the mixture component to be selected for each observation, are related through a Markov process rather than independent of each other. As shown in FIG. 5B, the HMM is typically defined by a set of hidden states, a matrix of state transition probabilities and a matrix of emission probabilities. Each hidden state has different statistical properties.
  • Mutations and genetic alterations including in copy number, for example, allelic imbalances, chromosomal copy number changes, such as amplifications, deletions, aneuploidy, loss of heterozygosity, and micro-satellite instability are often found to be associated with a disease state, for example, cancer. It has been observed that alterations in chromosomal copy number and loss of heterozygosity (LOH) are forms of genetic changes that often signal the activation of oncogenes and inactivation of tumor suppressor genes (anti-oncogenes). Variations in the form of copy number polymorphisms (CNP) can also occur in normal individuals. Identification of the loci implicated in these aberrations can generate anchor points which facilitate oncogenomics and toxicogenomics studies. Subsequently the shared LOH and aberrant CN regions can be used to partition the transcriptome data and track the differential transcript expression in the affected genomic segments. Locating and exploring such alteration events is an important research approach toward understanding the cause and progression of disease. For diploid organisms, the abnormal chromosomal state results when the normal diploid distribution is perturbed, resulting in changes that can include, for example, deletions, amplifications and translocations. Deletions can be of a partial chromosome ranging from micro-deletions on the order of several kb to macro-deletions of mega bases, entire arms of a chromosome or entire chromosomes. Amplifications can range from partial chromosomal amplifications to gains of a single copy of a chromosome to multiple copy gains of one or more chromosomes. Translocations generally comprise parts of a first chromosome being translocated to another chromosome.
  • FIG. 5B shows the general architecture of an instantiated HMM for mutation detection. Each oval shape X1, X2, X3 represents a random variable that can adopt any of a number of values. The random variable x(t) is the hidden state at time t (x(t)∈{x1, x2, x3}). The random variable y(t) is the observation at time t (with y(t)∈{y1, y2, y3, y4}). The arrows in the diagram (often called a trellis diagram) denote conditional dependencies. The conditional probability distribution of the hidden variable x(t) at time t, given the values of the hidden variable x at all times, depends only on the value of the hidden variablex (t−1): the values at time t−2 and before have no influence. This is called the Markov property. Similarly, the value of the observed variable y(t) representing the mutation conditions only depends on the value of the hidden variable x(t) (both at timet).
  • In FIG. 5B, the state space of the hidden variables is discrete, while the observations themselves can either be discrete (typically generated from a categorical distribution) or continuous (typically from a Gaussian distribution). The parameters of a hidden Markov model are of two types, transition probabilities and emission probabilities (also known as output probabilities). The transition probabilities control the way the hidden state at time is chosen given the hidden state at time. The hidden state space is assumed to consist of one of possible values, modeled as a categorical distribution. (See the section below on extensions for other possibilities.) This means that for each of the possible states that a hidden variable at time can be in, there is a transition probability from this state to each of the possible states of the hidden variable at time, for a total of transition probabilities. Note that the set of transition probabilities for transitions from any given state must sum to 1. Thus, the matrix of transition probabilities is a Markov matrix. Because any one transition probability can be determined once the others are known, there are a total of transition parameters.
  • In addition, for each of the possible states, there is a set of emission probabilities governing the distribution of the observed variable at a particular time given the state of the hidden variable at that time. The size of this set depends on the nature of the observed variable. For example, if the observed variable is discrete with possible values, governed by a categorical distribution, there will be separate parameters, for a total of emission parameters over all hidden states. On the other hand, if the observed variable is an -dimensional vector distributed according to an arbitrary multivariate Gaussian distribution, there will be parameters controlling the means and parameters controlling the covariance matrix, for a total of emission parameters. (In such a case, unless the value of is small, it may be more practical to restrict the nature of the covariances between individual elements of the observation vector, e.g. by assuming that the elements are independent of each other, or less restrictively, are independent of all but a fixed number of adjacent elements.).
  • The HMM method can model a somatic evolution of cancer. The method includes modeling genetic instability, which results in abnormal numbers of chromosomes or aneuploidy, elevated mutation rates, and altered distributions of mutational patterns.
  • The method can identify one or more cancer mutation drivers. These drivers include those that disrupt cellular signaling pathways essential for multicellular organisms and possible mutations that increase somatic fitness of cancer cells. The method can include identifying dynamics of tumor progression in a population based on interactions with an environment. The method includes collecting repeated genetic observations to enhance statistical inference about the evolution of tumors.
  • The method includes recommending or providing a therapeutic regimen in anticipation of the one or more escape paths. Diagnosis of cancer can be done by analyzing the genetic variants, even in the presence of noise. The analysis can be based on the frequency of Sequence Variants or Level of CNV and a diagnosis confidence indication or level for detecting genetic variants in the noise range can be established. The process increases the diagnosis confidence using a plurality of measurements to increase confidence of Diagnosis (6), or alternatively using measurements at a plurality of time points to determine whether cancer is advancing, in remission or stabilized. The diagnostic confidence can be used to identify disease states. For example, cell free polynucleotides taken from a subject can include polynucleotides derived from normal cells, as well as polynucleotides derived from diseased cells, such as cancer cells. Polynucleotides from cancer cells may bear genetic variants, such as somatic cell mutations and copy number variants. When cell free polynucleotides from a sample from a subject are sequenced, these cancer polynucleotides are detected as sequence variants or as copy number variants. The relative amount of tumor polynucleotides in a sample of cell free polynucleotides is referred to as the “tumor burden.” Measurements of a parameter, whether or not they are in the noise range, may be provided with a confidence interval. Tested over time, one can determine whether a cancer is advancing, stabilized or in remission by comparing confidence intervals over time. Where the confidence intervals do not overlap, this indicates the direction of disease.
  • In one implementation, using measurements from a plurality of samples collected substantially at once or over a plurality of time points, the diagnostic confidence indication for each variant can be adjusted to indicate a confidence of predicting the observation of the CNV or mutation. The confidence can be increased by using measurements at a plurality of time points to determine whether cancer is advancing, in remission or stabilized. The diagnostic confidence indication can be assigned by any of a number of known statistical methods is assigned and can be based, at least in part, on the frequency at which the measurements are observed over a period of time. For example, a statistical correlation of current and prior results can be done. Alternatively, for each diagnosis, a hidden Markov model can be built, such that a maximum likelihood or maximum a posteriori decision can be made based on the frequency of occurrence of a particular test event from a plurality of measurements or a time points. As part of this model, the probability of error and resultant diagnostic confidence indication for a particular decision can be output as well. In this manner, the measurements of a parameter, whether or not they are in the noise range, may be provided with a confidence interval. Tested over time, one can increase the predictive confidence of whether a cancer is advancing, stabilized or in remission by comparing confidence intervals over time. Two time points can be separated by about a month to about a year, about a year to about 5 years, or no more than about three months.
  • The HMM detect with high sensitivity genetic variation in a sample of initial genetic material. The methods involve using one to three of the following tools: First, the efficient conversion of individual polynucleotides in a sample of initial genetic material into sequence-ready tagged parent polynucleotides, so as to increase the probability that individual polynucleotides in a sample of initial genetic material will be represented in a sequence-ready sample. This can produce sequence information about more polynucleotides in the initial sample. Second, high yield generation of consensus sequences for tagged parent polynucleotides by high rate sampling of progeny polynucleotides amplified from the tagged parent polynucleotides, and collapsing of generated sequence reads into consensus sequences representing sequences of parent tagged polynucleotides. This can reduce noise introduced by amplification bias and/or sequencing errors, and can increase sensitivity of detection. Third, the noise in the detection of mutations and copy number variations is reduced by comparing prior sample analysis with the current sample and increasing a diagnostic confidence indication if the same mutations and copy number variations have appeared in prior analysis and otherwise decreasing the diagnostic confidence indication if this is the first time the sequence is observed.
  • FIG. 5C shows an exemplary model generated by the system of FIG. 2B for inferring tumor phylogeny from next-generation sequencing data. The subclones are related to each other by an evolutionary process of acquisition of mutations. In this example, the three clones (leaf nodes) are characterized by different combinations of the four single nucleotide variant (SNV) sets A, B, C, and D. The percentages on the edges of the tree indicate the fraction of cells with this particular set of SNVs, e.g., 70% of all cells carry A, 40% additionally carry B, and only 7% carry A, B, and D.
  • FIG. 5D shows an exemplary a heterogeneous collection of normal cells and cancer subclones developed during an evolutionary history of a tumor. The evolutionary history of a tumor gives rise to a heterogeneous collection of normal cells (small discs) and cancer subclones (large discs, triangles, squares). Internal nodes that have been fully replaced by their descendants (like the one carrying SNV sets A and B without C or D) are no longer part of the tumor.
  • Embodiments of the invention can take the form of an entirely hardware embodiment, an entirely software embodiment or an embodiment containing both hardware and software elements. In a preferred embodiment, the invention is implemented in software, which includes but is not limited to firmware, resident software, microcode, and the like. Furthermore, the invention can take the form of a computer program product accessible from a computer-usable or computer-readable medium providing program code for use by or in connection with a computer or any instruction execution system.
  • For the purposes of this description, a computer-usable or computer readable medium can be any apparatus that can contain, store, communicate, or transport the program for use by or in connection with the instruction execution system, apparatus, or device. The medium can be an electronic, magnetic, optical, electromagnetic, infrared, or semiconductor system (or apparatus or device). Examples of a computer-readable medium include a semiconductor or solid state memory, magnetic tape, a removable computer diskette, a random access memory (RAM), a read-only memory (ROM), a rigid magnetic disk and an optical disk. Current examples of optical disks include compact disk-read only memory (CD-ROM), compact disk-read/write (CD-R/W) and DVD.
  • A data processing system suitable for storing and/or executing program code will include at least one processor coupled directly or indirectly to memory elements through a system bus. The memory elements can include local memory employed during actual execution of the program code, bulk storage, and cache memories which provide temporary storage of at least some program code in order to reduce the number of times code must be retrieved from bulk storage during execution. Input/output or I/O devices (including but not limited to keyboards, displays, pointing devices, etc.) can be coupled to the system either directly or through intervening I/O controllers. Network adapters may also be coupled to the system to enable the data processing system to become coupled to other data processing systems or remote printers or storage devices through intervening private or public networks. Modems, cable modem and Ethernet cards are just a few of the currently available types of network adapters.
  • APPENDIX
  • Drug Therapeutic Area Biomarker Referenced Subgroup Labeling Sections
    Abacavir Infectious HLA-B HLA-B*5701 allele Boxed Warning,
    Diseases carriers Contraindications,
    Warnings and
    Precautions
    Pharmacogenomic Oncology ERBB2 HER2 protein Indications and
    Information Ado- overexpression or gene Usage, Warnings and
    Trastuzumab amplification positive Precautions, Adverse
    Emtansine Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Pharmacogenomic Oncology EGFR EGFR exon 19 deletion Indications and
    Information on or exon 21 substitution Usage, Dosage and
    Afatinib (L858R)positive Administration,
    Adverse Reactions,
    Clinical
    Pharmacology,
    Clinical Studies
    Amitriptyline Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Anastrozole Oncology ESR1, PGR Hormone receptor- Indications and
    positive Usage, Adverse
    Reactions, Drug
    Interactions, Clinical
    Studies
    Arformoterol (1) Pulmonary UGT1A1 UGT1A1 poor Clinical
    metabolizers Pharmacology
    Arformoterol (2) Pulmonary CYP2D6 CYP2D6 intermediate Clinical
    or poor metabolizers Pharmacology
    Aripiprazole Psychiatry CYP2D6 CYP2D6 poor Dosage and
    metabolizers Administration,
    Clinical
    Pharmacology
    Pharmacogenomic Oncology PML-RARA PML-RARα Clinical
    Information on translocation positive Pharmacology,
    Arsenic Trioxide Indications and
    Usage
    Atomoxetine Psychiatry CYP2D6 CYP2D6 poor Dosage and
    metabolizers Administration,
    Warnings and
    Precautions, Drug
    Interactions, Clinical
    Pharmacology
    Azathioprine Rheumatology TPMT TPMT intermediate or Clinical
    poor metabolizers Pharmacology,
    Warnings,
    Precautions Drug
    Interactions, Adverse
    Reactions, Dosage
    and Administration
    Boceprevir Infectious IFNL3 IL28B rs12979860 T Clinical
    Diseases allele carriers (C/T and Pharmacology
    T/T genotype)
    Bosutinib Oncology BCR/ABL1 Philadelphia Indications and
    chromosome positive Usage Adverse
    Reactions, Use in
    Specific Populations,
    Clinical Studies
    Busulfan Oncology BCR-ABL1 Philadelphia Clinical Studies
    chromosome negative
    Capecitabine Oncology DPYD DPD deficient Contraindications,
    Warnings and
    Precautions, Patient
    Information
    Carbamazepine (1) Neurology HLA-B HLA-B*1502 allele Boxed Warning,
    carriers Warnings,
    Precautions
    Carbamazepine (2) Neurology HLA-A HLA-A*3101 allele Warnings
    carriers
    Pharmacogenomic Inborn Errors of NAGS N-acetylglutamate Indications and
    Information on Metabolism synthase deficient Usage, Warnings and
    Carglumic Acid Precautions, Use in
    Specific Populations
    Clinical
    Pharmacology
    Clinical Studies
    Carisoprodol Rheumatology CYP2C19 CYP2C19 poor Use in Specific
    metabolizers Populations, Clinical
    Pharmacology
    Carvedilol Cardiology CYP2D6 CYP2D6 poor Drug Interactions,
    metabolizers Clinical
    Pharmacology
    Celecoxib Rheumatology CYP2C9 CYP2C9 poor Dosage and
    metabolizers Administration, Use
    in Specific
    Populations, Clinical
    Pharmacology
    Pharmacogenomic Oncology ALK ALK gene Indications and
    Information on rearrangement positive Usage, Adverse
    Ceritinib Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Cetuximab (1) Oncology EGFR EGFR protein Indications and
    expression positive Usage, Dosage and
    Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Cetuximab (2) Oncology KRAS KRAS codon 12 and 13 Indications and
    mutation negative Usage, Dosage and
    Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Cevimeline Dental CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Chloroquine Infectious G6PD G6PD deficient Precautions
    Diseases
    Chlorpropamide Endocrinology G6PD G6PD deficient Precautions
    Cisplatin Oncology TPMT TPMT intermediate or Clinical
    poor metabolizers Pharmacology,
    Warnings,
    Precautions, Adverse
    Reactions
    Citalopram (1) Psychiatry CYP2C19 CYP2C19 poor Clinical
    metabolizers Pharmacology,
    Warnings, Dosage
    and Administration
    Citalopram (2) Psychiatry CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology
    Clobazam Neurology CYP2C19 CYP2C19 poor Dosage and
    metabolizers Administration, Use
    in Specific
    Populations, Clinical
    Pharmacology
    Clomipramine Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Clopidogrel Cardiology CYP2C19 CYP2C19 intermediate Boxed Warning,
    or poor metabolizers Dosage and
    Administration,
    Warnings and
    Precautions, Clinical
    Pharmacology
    Clozapine Psychiatry CYP2D6 CYP2D6 poor Dosage and
    metabolizers Administration, Use
    in Specific
    Populations, Clinical
    Pharmacology
    Codeine Anesthesiology CYP2D6 CYP2D6 ultra-rapid Boxed Warnings,
    metabolizers Warnings and
    Precautions, Use in
    Specific Populations,
    Patient Counseling
    Information
    Pharmacogenomic Oncology ALK ALK gene Indications and
    Information on rearrangement positive Usage, Dosage and
    Crizotinib Administration,
    Adverse Reactions,
    Clinical
    Pharmacology,
    Clinical Studies
    Dabrafenib (1) Oncology BRAF BRAF V600E/K Indications and
    mutation positive Usage, Dosage and
    Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Clinical
    Pharmacology,
    Clinical Studies,
    Patient Counseling
    Information
    Dabrafenib (2) Oncology G6PD G6PD deficient Warnings and
    Precautions, Adverse
    Reactions, Patient
    Counseling
    Information
    Dapsone (1) Dermatology G6PD G6PD deficient Warnings and
    Precautions, Use in
    Specific Populations,
    Patient Counseling
    Information
    Dapsone (2) Infectious G6PD G6PD deficient Precautions, Adverse
    Diseases Reactions,
    Overdosage
    Pharmacogenomic Oncology BCR/ABL1 Philadelphia Indications and
    Information on chromosome positive;, Usage, Dosage and
    Dasatinib T315I mutation-positive Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Pharmacogenomic Oncology IL2RA CD25 antigen positive Indications and
    Information on Usage, Warnings and
    Denileukin Diftitox Precautions, Clinical
    Studies
    Desipramine Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Dexlansoprazole Gastroenterology CYP2C19 CYP2C19 poor Drug Interactions,
    metabolizers Clinical
    Pharmacology
    Dextromethorphan Neurology CYP2D6 CYP2D6 poor Warnings and
    and Quinidine metabolizers Precautions, Clinical
    Pharmacology
    Diazepam Psychiatry CYP2C19 CYP2C19 poor Clinical
    metabolizers Pharmacology
    Doxepin (1) Psychiatry CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology
    Doxepin (2) Psychiatry CYP2C19 CYP2C19 poor Clinical
    metabolizers Pharmacology
    Drospirenone and Gynecology, CYP2C19 CYP2C19 intermediate Clinical
    Ethinyl Estradiol Dermatology metabolizers Pharmacology
    Eliglustat Inborn Errors of CYP2D6 CYP2D6 ultrarapid, Indications and
    Metabolism intermediate or poor Usage, Dosage and
    metabolizers Administration,
    Contraindications,
    Warnings and
    Precautions, Drug
    Interactions, Use in
    Specific Populations,
    Clinical
    Pharmacology,
    Clinical Studies
    Eltrombopag (1) Hematology F5 Factor V Leiden carriers Warnings and
    Precautions
    Eltrombopag (2) Hematology SERPINC1 Antithrombin III Warnings and
    deficient Precautions
    Erlotinib (1) Oncology EGFR EGFR protein Clinical Studies
    expression positive
    Erlotinib (2) Oncology EGFR EGFR exon 19 deletion Indications and
    or exon 21 substitution Usage Dosage and
    (L858R) positive Administration,
    Adverse Reactions,
    Clinical
    Pharmacology
    Clinical Studies
    Esomeprazole Gastroenterology CYP2C19 CYP2C19 poor Drug Interactions,
    metabolizers Clinical
    Pharmacology
    Everolimus (1) Oncology ERBB2 HER2 protein Indications and
    overexpression negative Usage, Dosage and
    Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Drug
    Interactions, Use in
    Specific Populations,
    Clinical
    Pharmacology,
    Clinical Studies
    Everolimus (2) Oncology ESR1 Estrogen receptor Clinical Studies
    positive
    Exemestane (1) Oncology ESR1 Estrogen receptor Indications and
    positive Usage, Dosage and
    Administration,
    Clinical Studies
    Exemestane (2) Oncology PGR Progesterone receptor Clinical Studies
    positive
    Fluorouracil (1) Dermatology DPYD DPD deficient Contraindications,
    Warnings, Patient
    Information
    Fluorouracil (2) Oncology DPYD DPD deficient Warnings
    Fluoxetine Psychiatry CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology,
    Warnings,
    Precautions
    Flurbiprofen Rheumatology CYP2C9 CYP2C9 poor Clinical
    metabolizers Pharmacology
    Fluvoxamine Psychiatry CYP2D6 CYP2D6 poor Drug Interactions
    metabolizers
    Fulvestrant Oncology ESR1, PGR Hormone receptor Indications and
    positive Usage, Clinical
    Pharmacology,
    Clinical Studies
    Galantamine Neurology CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology
    Glimepiride Endocrinology G6PD G6PD deficient Warning and
    Precautions, Adverse
    Reactions
    Glipizide Endocrinology G6PD G6PD deficient Precautions
    Glyburide Endocrinology G6PD G6PD deficient Precautions
    Pharmacogenomic Oncology del (17p) Chromosome 17p Indications and
    Information on deletion positive Usage, Clinical
    Ibrutinib Studies
    Iloperidone Psychiatry CYP2D6 CYP2D6 poor Dosage and
    metabolizers Administration,
    Warnings and
    Precautions, Drug
    Interactions, Clinical
    Pharmacology
    Imatinib (1) Oncology KIT KIT protein expression Indications and
    positive, c-KIT D816V Usage, Dosage and
    mutation negative Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Use in
    Specific Populations,
    Clinical
    Pharmacology,
    Clinical Studies
    Imatinib (2) Oncology BCR-ABL1 Philadelphia Indications and
    chromosome positive Usage, Dosage and
    Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Use in
    Specific Populations,
    Clinical
    Pharmacology,
    Clinical Studies
    Imatinib (3) Oncology PDGFRB PDGFR gene Indications and
    rearrangement positive Usage, Dosage and
    Administration,
    Clincal Studies
    Imatinib (4) Oncology FIP1L1- FIP1L1-PDGFRα fusion Indications and
    PDGFRA kinase (or CHIC2 Usage, Dosage and
    deletion) positive Administration,
    Clinical Studies
    Imipramine Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Indacaterol Pulmonary UGT1A1 UGT1A1-*28 allele Clinical
    homozygotes Pharmacology
    Irinotecan Oncology UGT1A1 UGT1A1*28 allele Dosage and
    carriers Administration,
    Warnings and
    Precautions, Clinical
    Pharmacology
    Isosorbide and Cardiology NAT1-2 Slow acetylators Clinical
    Hydralazine Pharmacology
    Ivacaftor Pulmonary CFTR CFTR G551D, G1244E, Indications and
    G1349D, G178R, Usage, Adverse
    G551S, S1251N, Reactions, Use in
    S1255P, S549N, or Specific Populations,
    S549R mutation Clinical
    carriers, F508del Pharmacology,
    mutation homozygotes Clinical Studies
    Lansoprazole Gastroenterology CYP2C19 CYP2C19 intermediate Drug Interactions
    or poor metabolizers
    Lapatinib (1) Oncology ERBB2 HER2 protein Indications and
    overexpression positive Usage, Dosage and
    Administration,
    Adverse Reactions,
    Use in Specific
    Populations, Clinical
    Pharmacology,
    Clinical Studies
    Lapatinib (2) Oncology HLA-DQA1, HLA-DQA1*0201 or Clinical
    HLA-DRB1 -DRB1*0701 allele Pharmacology
    carriers
    Pharmacogenomic Hematology del (5q) Chromosome 5q Boxed Warning,
    Information on deletion positive Indications and
    Lenalidomide Usage, Adverse
    Reactions, Use in
    Specific Populations,
    Clinical Studies
    Letrozole Oncology ESR1, PGR Hormone receptor Indications and
    positive Usage, Adverse
    Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Lomitapide Endocrinology LDLR LDLR mutation Indication and Usage,
    homozygotes Warnings and
    (homozygous familial Precautions, Adverse
    hypercholesterolemia) Reactions, Clinical
    Studies
    Mafenide Infectious G6PD G6PD deficient Warnings, Adverse
    Diseases Reactions
    Pharmacogenomic Oncology TPMT TPMT intermediate or Clinical
    Information on poor metabolizers Pharmacology,
    Mercaptopurine Warnings,
    Precautions, Adverse
    Reactions, Dosage
    and Administration
    Methylene Blue Hematology G6PD G6PD deficient Precautions
    Metoclopramide Gastroentrology CYB5R1-4 NADH cytochrome b5 Precautions
    reductase deficient
    Metoprolol Cardiology CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology
    Mipomersen Endocrinology LDLR LDLR mutation Boxed Warning,
    heterozygotes and Indications and
    homozygotes Usage, Warnings and
    (heterozygous and Precautions, Adverse
    homozygous familial Reactions, Use in
    hypercholesterolemia) Specific Populations,
    Clinical Studies
    Modafinil Psychiatry CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology,
    Precautions
    Mycophenolic Acid Transplantation HPRT1 HGPRT deficient Warnings and
    Precautions
    Nalidixic Acid Infectious G6PD G6PD deficient Precautions, Adverse
    Diseases Reactions
    Nefazodone Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Nilotinib (1) Oncology BCR-ABL Philadelphia Indications and
    chromosome positive Usage, Dosage and
    Administration,
    Adverse Reactions,
    Use in Specififc
    Populations, Clinical
    Pharmacology,
    Clinical Studies
    Nilotinib (2) Oncology UGT1A1 UGT1A1*28 allele Clinical
    homozygotes Pharmacology
    Nitrofurantoin Infectious G6PD G6PD deficient Warnings, Adverse
    Diseases Reactions
    Nortriptyline Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Obinutuzumab Oncology MS4A1 CD20 antigen positive Clinical Studies
    Omacetaxine Oncology BCR-ABL1 Philadelphia Clinical
    chromosome positive Pharmacology,
    Clinical Studies
    Omeprazole Gastroenterology CYP2C19 CYP2C19 poor Drug Interactions
    metabolizers
    Panitumumab (1) Oncology EGFR EGFR protein Clinical
    expression positive Pharmacology,
    Clinical Studies
    Panitumumab (2) Oncology KRAS KRAS codon 12 and 13 Indications and
    mutation negative Usage, Dosage and
    Administration,
    Warnings and
    Precautions, Adverse
    Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Pantoprazole Gastroenterology CYP2C19 CYP2C19 poor Clinical
    metabolizers Pharmacology
    Paroxetine Psychiatry CYP2D6 CYP2D6 extensive Drug Interactions
    metabolizers
    Pharmacogenomic Oncology UGT1A1 UGT1A1*28 allele Clinical
    Information on homozygotes Pharmacology,
    Pazopanib (TA)7/(TA)7 genotype Warnings and
    Precautions
    PEG-3350, Sodium Gastroenterology G6PD G6PD deficient Warnings and
    Sulfate, Sodium Precautions
    Chloride, Potassium
    Chloride, Sodium
    Ascorbate, and
    Ascorbic Acid
    Pegloticase Rheumatology G6PD G6PD deficient Contraindications,
    Patient Counseling
    Information
    Perphenazine Psychiatry CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology,
    Precautions
    Pharmacogenomic Oncology ERBB2 HER2 protein Indications and
    Information overexpression positive Usage, Warnings and
    onPertuzumab Precautions, Adverse
    Reactions, Clinical
    Pharmacology,
    Clinical Studies
    Phenytoin Neurology HLA-B HLA-B*1502 allele Warnings
    carriers
    Pimozide Psychiatry CYP2D6 CYP2D6 poor Precautions, Dosage
    metabolizers and Administration
    Pharmacogenomic Oncology BCR-ABL1 Philadelphia Indications and
    Information on chromosome positive; Usage, Warnings and
    Ponatinib T315I mutation positive Precautions, Adverse
    Reactions, Use in
    Specific Populations,
    Clinical
    Pharmacology,
    Clinical Studies
    Prasugrel (1) Cardiology CYP2C19 CYP2C19 poor Use in Specific
    metabolizers Populations, Clinical
    Pharmacology,
    Clinical Studies
    Prasugrel (2) Cardiology CYP2C9 CYP2C9 variant carriers Use in Specific
    Populations, Clinical
    Pharmacology,
    Clinical Studies
    Prasugrel (3) Cardiology CYP3A5 CYP3A5 variant Use in Specific
    carriers Populations, Clinical
    Pharmacology,
    Clinical Studies
    Prasugrel (4) Cardiology CYP2B6 CYP2B6 variant carriers Use in Specific
    Populations, Clinical
    Pharmacology,
    Clinical Studies
    Pravastatin Endocrinology LDLR LDLR mutation Indications and
    heterozygotes and Usage, Use in
    homozygotes Specific Populations,
    (heterozygous and Clinical Studies
    homozygous familial
    hypercholesterolemia)
    Primaquine Infectious G6PD G6PD deficient Warnings and
    Diseases Precautions, Adverse
    Reactions
    Propafenone Cardiology CYP2D6 CYP2D6 poor Dosage and
    metabolizers Administration,
    Warnings and
    Precautions, Clinical
    Pharmacology
    Propranolol Cardiology CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology
    Protriptyline Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Quinidine Cardiology CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Quinine Sulfate (1) Infectious G6PD G6PD deficient Contraindications
    Diseases
    Quinine Sulfate (2) Infectious CYP2D6 CYP2D6 poor Drug Interactions
    Diseases metabolizers
    Rabeprazole Gastroenterology CYP2C19 CYP2C19 poor Drug Interactions,
    metabolizers Clinical
    Pharmacology
    Rasburicase (1) Oncology G6PD G6PD deficient Boxed Warning,
    Contraindications,
    Warnings and
    Precautions
    Rasburicase (2) Oncology CYB5R1-4 NADH cytochrome b5 Warnings and
    reductase deficient Precautions
    Rifampin, Isoniazid, Infectious NAT1-2 Slow acetylators Clinical
    and Pyrazinamide Diseases (inactivators) Pharmacology,
    Adverse Reactions
    Risperidone Psychiatry CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology
    Pharmacogenomic Oncology MS4A1 CD20 antigen positive Indications and
    Information Usage, Dosage and
    onRituximab Administration,
    Adverse Reactions,
    Use in Specific
    Populations, Clinical
    Pharmacology,
    Clinical Studies
    Simeprevir Infectious IFNL3 IL28B rs12979860 T Clinical
    Diseases allele carriers Pharmacology,
    Clinical Studies
    Sodium Nitrite Toxicology G6PD G6PD deficient Warnings and
    Precautions
    Sodium Inborn Errors of NAGS Urea cycle enzyme Indications and
    phenylacetate and Metabolism CPS1, deficient Usage, Dosage and
    Sodium Benzoate ASS1, OTC, Administration,
    ASL, ABL2 Warnings and
    Precautions, Adverse
    Reactions, Drug
    Interactions, Use in
    Specific Populations,
    Overdosage, Clinical
    Pharmacology,
    Clinical Studies
    Sofosbuvir Infectious IFNL3 IL28B rs12979860 T Clinical Studies
    Diseases allele carriers (non-C/C
    genotype)IL28B
    Succimer Hematology G6PD G6PD deficient Clinical
    Pharmacology
    Sulfamethoxazole Infectious G6PD G6PD deficient Precautions
    and Trimethoprim Diseases
    Tamoxifen (1) Oncology ESR1, PGR Hormone receptor Clinical
    positive Pharmacology,
    Indications and
    Usage, Precautions,
    Adverse Reactions,
    Medication Guide
    Tamoxifen (2) Oncology F5 Factor V Leiden carriers Warnings
    Tamoxifen (3) Oncology F2 Prothrombin 20210A Warnings
    allele positive
    Telaprevir Infectious IFNL3 IL28B rs12979860 T Clinical
    Diseases allele carriers (C/T and Pharmacology,
    T/T genotype) Clinical Studies
    Tetrabenazine Neurology CYP2D6 CYP2D6 poor Dosage and
    metabolizers Administration,
    Warnings and
    Precautions, Use in
    Specific Populations,
    Clinical
    Pharmacology
    Thioguanine Oncology TPMT TPMT intermediate or Warnings,
    poor metabolizers Precautions, Dosage
    (deficient) and Administration
    Thioridazine Psychiatry CYP2D6 CYP2D6 poor Contraindications,
    metabolizers Warnings,
    Precautions
    Ticagrelor Cardiology CYP2C19 CYP2C19 poor Clinical Studies
    metabolizers
    Tolterodine Genitourinary CYP2D6 CYP2D6 poor Warnings and
    metabolizers Precautions, Drug
    Interactions, Use in
    Specific Populations,
    Clinical
    Pharmacology
    Tositumomab Oncology MS4A1 CD20 antigen positive Indications and
    Usage, Clinical
    Pharmacology
    Tramadol Analgesic CYP2D6 CYP2D6 poor Clinical
    metabolizers Pharmacology
    Pharmacogenomic Oncology BRAF BRAF V600E/K Indications and
    Information mutation positive Usage, Dosage and
    onTrametinib Administration,
    Adverse Reactions,
    Clinical
    Pharmacology,
    Clinical Studies,
    Patient Counseling
    Information
    Pharmacogenomic Oncology ERBB2 HER2 protein Indications and
    Information on overexpression positive Usage, Warnings and
    Trastuzumab Precautions, Clinical
    Pharmacology,
    Clinical Studies
    Tretinoin Oncology PML/RARA PML/RARα Clinical
    translocation positive Pharmacology,
    Indications and
    Usage, Warnings
    Trimipramine Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Valproic Acid (1) Neurology POLG POLG mutation positive Boxed Warning,
    Contraindications,
    Warnings and
    Precautions
    Valproic Acid (2) Neurology NAGS, Urea cycle enzyme Contraindications,
    CPS1, deficient Warnings and
    ASS1, OTC, Precautions
    ASL, ABL2
    Pharmacogenomic Oncology BRAF BRAF V600E mutation Indications and
    Information on positive Usage, Dosage and
    Vemurafenib Administration,
    Warning and
    Precautions, Clinical
    Pharmacology,
    Clinical Studies,
    Patient Counseling
    Information
    Venlafaxine Psychiatry CYP2D6 CYP2D6 poor Precautions
    metabolizers
    Voriconazole Infectious CYP2C19 CYP2C19 intermediate Clinical
    Diseases or poor metabolizers Pharmacology
    Vortioxetine Neurology CYP2D6 CYP2D6 poor Dosage and
    metabolizers Administration,
    Clinical
    Pharmacology
    Warfarin (1) Cardiology or CYP2C9 CYP2C9 intermediate Dosage and
    Hematology or poor metabolizers Administration, Drug
    Interactions, Clinical
    Pharmacology
    Warfarin (2) Cardiology or VKORC1 VKORC1 A allele Dosage and
    Hematology carriers Administration,
    Clinical
    Pharmacology
    Warfarin (3) Cardiology, PROS Protein S deficient Warnings and
    Hematology Precautions
    Warthrin (4) Cardiology, PROC Protein C deficient Warnings and
    Hematology Precautions

Claims (20)

What is claimed is:
1. A system to provide information on a drug substance for a subject, comprising:
a network unit to receive gene or DNA sequencing data; and
a processor running:
code to match genomic biomarker(s) from gene or DNA sequencing for a population with historical information for a population on drug structure, dosage, clinical variability and risk for adverse events for the drug substance, the computer constructing side effect features for each drug, and applying a classifier to the features to predict one or more adverse drug interactions, the computer generating one or more indicia for the drug substance; and
code to apply the indicia to the subject DNA to provide personalized medicine.
2. The system of claim 1, wherein the biomarkers comprise at least one of: germ-line or somatic gene variants, functional deficiencies, expression changes, chromosomal abnormalities, and protein biomarkers used to select patients for treatment.
3. The system of claim 1, comprising indicia on gene, protein or chromosomal testing, genetic testing, functional protein assays, cytogenetic studies before using the drug and an indicia about changes in efficacy, dosage or toxicity due to genetic variants, an indicia on a gene or protein involved in the metabolism or pharmacodynamics of the drug, and an indicia that the gene or protein leads to different response.
4. The system of claim 1, comprising a code communicating with: a gene-drug interactions training dataset that includes pharmaceutical and pharmacogenomics interactions for the drug; a side effect feature database for the drug from gene sensitivity associated with the drug; a gene-drug interactions classifier that predicts adverse gene-drug interactions for drug gene pairs and a subject's genetic scan; and for each of the side effects, a Fisher's exact test to determine predicted gene-drug-drug interactions.
5. The system of claim 1, wherein the substance is a drug, comprising code for: constructing a gene-drug interactions training dataset that includes pharmaceutical, pharmacokinetic or pharmacodynamics, and pharmacogenomics drug-drug interactions for each drug; constructing side effect features for each of the plurality of drugs from side effects associated with the plurality of drugs; running a gene-drug-drug interactions classifier that predicts adverse drug-drug interactions for drug pairs and the genetic scan; and for each of the side effects, performing a Fisher's exact test to determine predicted gene-drug-drug interactions.
6. The system of claim 5, wherein the gene-drug-drug interactions classifier uses a classifying objective function having a smoothness constraint and a fitting constraint to render the predicted adverse gene-drug interactions.
7. The system of claim 1, comprising code for building a genetic drug-drug interactions classifier using deep learning with a neural network, comprising propagating drug-drug interactions between different ones of the plurality of drugs on a basis that if a first drug has an interaction with a second drug and the second drug is similar to a third drug, then the first drug is considered as having interaction with the third drug.
8. The system of claim 1, wherein the indicia comprise a bar code, a near field communication (NFC) transmission, a text, or an Internet link.
9. The system of claim 1, wherein the indicia is readable by a smart phone, comprising code to update the indicia with changes in the subject after the consumption of the substance.
10. The system of claim 1, comprising a gene sequencer to capture genetic data from the subject, wherein the indicia is updated with changes in the subject after the consumption of the substance.
11. The system of claim 1, comprising a module detect the evolutionary paths of escape
12. The system of claim 1, comprising a hidden markov model (HMM) to detect evolutionary paths of escape.
13. The system of claim 1, comprising a module to detect mutation by comparing later sequence reads by a genetic analyzer with prior sequence reads.
14. The system of claim 1, comprising a module to detect an amount of mutation cancer polynucleotides in a sample from a subject over time by determining a frequency of the cancer polynucleotides at a plurality of time points; determining an error range for the frequency at each of the plurality of time points; determining, between an earlier and later time point, whether error ranges (1) overlap, indicating stability of frequency, (2) an increase at the later time point outside the error range, indicating increase in frequency or (3) a decrease at the later time point outside the error range, indicating decrease in frequency.
15. The system of claim 1, comprising a model to infer tumor phylogeny from sequencing data, wherein subclones are related to each other by an evolutionary process of acquisition of mutations.
16. The system of claim 1, comprising a deep learning machine using deep convolutionary neural networks for detecting genetic based drug-drug interaction.
17. The system of claim 1, comprising code to:
acquire subject genetic scans using a gene sequencer;
identify each substance to be provided to the subject;
determine substance interactions for each drug and the genetic scan; and
provide indicia associated with each substance to warn the subject or a medical provider based on the genetic scan.
18. The system of claim 1, comprising a module for analyzing a disease state of a subject by collecting genetic profile data on a population of tumors and original tumor treatment(s); identifying one or more evolutionary paths of escape and evolved tumor treatment(s); and based on a subject profile, predicting a probability of escape along the one or more evolutionary paths.
19. The system of claim 1, comprising a module to recommend drug repurposing using drug-effect similarities from existing therapeutic indications of known drugs.
20. The system of claim 1, comprising a module to predict side effects before a drug enters a clinical trial.
US16/267,651 2013-03-15 2019-02-05 Pharmacogenetic drug interaction management system Abandoned US20190172588A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/267,651 US20190172588A1 (en) 2013-03-15 2019-02-05 Pharmacogenetic drug interaction management system

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US13/843,903 US20140278475A1 (en) 2013-03-15 2013-03-15 Tele-analytics based treatment recommendations
US15/090,466 US10262107B1 (en) 2013-03-15 2016-04-04 Pharmacogenetic drug interaction management system
US16/267,651 US20190172588A1 (en) 2013-03-15 2019-02-05 Pharmacogenetic drug interaction management system

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/090,466 Continuation US10262107B1 (en) 2013-03-15 2016-04-04 Pharmacogenetic drug interaction management system

Publications (1)

Publication Number Publication Date
US20190172588A1 true US20190172588A1 (en) 2019-06-06

Family

ID=66098724

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/090,466 Expired - Fee Related US10262107B1 (en) 2013-03-15 2016-04-04 Pharmacogenetic drug interaction management system
US16/267,651 Abandoned US20190172588A1 (en) 2013-03-15 2019-02-05 Pharmacogenetic drug interaction management system

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/090,466 Expired - Fee Related US10262107B1 (en) 2013-03-15 2016-04-04 Pharmacogenetic drug interaction management system

Country Status (1)

Country Link
US (2) US10262107B1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200119986A1 (en) * 2017-11-03 2020-04-16 Vignet Incorporated Systems and methods for managing operation of devices in complex systems and changing environments
WO2021211881A1 (en) * 2020-04-17 2021-10-21 Cipherome, Inc. Prediction of adverse drug reaction based on machine-learned models using protein function scores and clinical factors
US20210327543A1 (en) * 2018-07-27 2021-10-21 Karydo Therapeutix, Inc. Artificial Intelligence Model for Predicting Actions of Test Substance in Humans
US11158423B2 (en) 2018-10-26 2021-10-26 Vignet Incorporated Adapted digital therapeutic plans based on biomarkers
US11238979B1 (en) 2019-02-01 2022-02-01 Vignet Incorporated Digital biomarkers for health research, digital therapeautics, and precision medicine
US11281553B1 (en) 2021-04-16 2022-03-22 Vignet Incorporated Digital systems for enrolling participants in health research and decentralized clinical trials
US11302448B1 (en) 2020-08-05 2022-04-12 Vignet Incorporated Machine learning to select digital therapeutics
US11322260B1 (en) 2020-08-05 2022-05-03 Vignet Incorporated Using predictive models to predict disease onset and select pharmaceuticals
US11456080B1 (en) 2020-08-05 2022-09-27 Vignet Incorporated Adjusting disease data collection to provide high-quality health data to meet needs of different communities
US11482305B2 (en) 2018-08-18 2022-10-25 Synkrino Biotherapeutics, Inc. Artificial intelligence analysis of RNA transcriptome for drug discovery
US11504011B1 (en) 2020-08-05 2022-11-22 Vignet Incorporated Early detection and prevention of infectious disease transmission using location data and geofencing
US11586524B1 (en) 2021-04-16 2023-02-21 Vignet Incorporated Assisting researchers to identify opportunities for new sub-studies in digital health research and decentralized clinical trials
US11705230B1 (en) 2021-11-30 2023-07-18 Vignet Incorporated Assessing health risks using genetic, epigenetic, and phenotypic data sources
US11789837B1 (en) 2021-02-03 2023-10-17 Vignet Incorporated Adaptive data collection in clinical trials to increase the likelihood of on-time completion of a trial
WO2023250312A1 (en) * 2022-06-21 2023-12-28 Parata Systems, Llc Methods, systems, and computer program product for pre-categorizing drug products based on characteristics thereof to reduce artificial intelligence model size used in validating the drug products
US11901083B1 (en) 2021-11-30 2024-02-13 Vignet Incorporated Using genetic and phenotypic data sets for drug discovery clinical trials

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9836582B2 (en) * 2008-08-14 2017-12-05 Srr Patent Holdings, Llc Drug substance interaction and generic substance information retrieval
US10679004B2 (en) * 2016-08-15 2020-06-09 International Business Machines Corporation Performing chemical textual analysis
US20180330808A1 (en) * 2017-05-10 2018-11-15 Petuum Inc. Machine learning system for disease, patient, and drug co-embedding, and multi-drug recommendation
WO2019067567A1 (en) * 2017-09-26 2019-04-04 Juno Diagnostics, Inc. Devices, systems and methods for biomarker analysis
US10803581B2 (en) * 2017-11-06 2020-10-13 Beijing Keya Medical Technology Co., Ltd. System and method for generating and editing diagnosis reports based on medical images
US20200027556A1 (en) 2018-07-17 2020-01-23 Petuum Inc. Systems and Methods for Medical Topic Discovery Based on Large-Scale Machine Learning
US11961605B2 (en) 2019-11-29 2024-04-16 Kpn Innovations, Llc Methods and systems for informing self-treatment remedy selection
US11443854B2 (en) * 2020-02-24 2022-09-13 International Business Machines Corporation Identifying potential medicinal interactions for online clinical trial study groups
CN111477287B (en) * 2020-06-28 2020-11-20 天云融创数据科技(北京)有限公司 Drug target prediction method, device, equipment and medium
CN111933302B (en) * 2020-10-09 2021-01-05 平安科技(深圳)有限公司 Medicine recommendation method and device, computer equipment and storage medium
CN112768089B (en) * 2021-04-09 2021-06-22 至本医疗科技(上海)有限公司 Method, apparatus and storage medium for predicting drug sensitivity status
CN112863696B (en) * 2021-04-25 2021-09-07 浙江大学 Drug sensitivity prediction method and device based on transfer learning and graph neural network
CN112989207B (en) * 2021-04-27 2021-08-27 武汉卓尔数字传媒科技有限公司 Information recommendation method and device, electronic equipment and storage medium
CN114373550B (en) * 2022-03-21 2022-06-21 普瑞基准科技(北京)有限公司 Medicine IC50 deep learning model prediction method based on molecular structure and gene expression

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7196071B2 (en) * 2000-09-20 2007-03-27 The General Hospital Corporation Methods of decreasing or preventing pain using spicamycin derivatives
US20050208512A1 (en) * 2003-10-01 2005-09-22 The Ohio State University Research Foundation Determining the chemosensitivity of cells to cytotoxic agents
JP4979382B2 (en) * 2003-10-06 2012-07-18 ノバルティス アーゲー Use of genetic polymorphisms to predict drug-induced hepatotoxicity
WO2006020126A2 (en) * 2004-07-14 2006-02-23 Protometrix, Inc. Method for providing protein microarrays
US8261058B2 (en) * 2005-03-16 2012-09-04 Dt Labs, Llc System, method and apparatus for electronically protecting data and digital content
US8151835B2 (en) * 2006-08-23 2012-04-10 Fht, Inc. Automated drug delivery bag filling system
US20100073202A1 (en) * 2008-09-25 2010-03-25 Mazed Mohammad A Portable internet appliance
WO2008083313A2 (en) * 2007-01-01 2008-07-10 Medrad, Inc. Methods and systems for integrated radiopharmaceutical generation, preparation, transportation, and administration
US8212677B2 (en) * 2007-02-12 2012-07-03 Alexander Ferguson Automated medication management system and method for use
US20130028480A1 (en) * 2008-08-14 2013-01-31 Srr Patent Holdings, Llc Generic substance information retrieval using mobile device
GB201115099D0 (en) * 2011-09-01 2011-10-19 Belgian Volition Sa Method for detecting nucleosomes
US10191929B2 (en) * 2013-05-29 2019-01-29 Noblis, Inc. Systems and methods for SNP analysis and genome sequencing

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11616688B1 (en) 2017-11-03 2023-03-28 Vignet Incorporated Adapting delivery of digital therapeutics for precision medicine
US10938651B2 (en) * 2017-11-03 2021-03-02 Vignet Incorporated Reducing medication side effects using digital therapeutics
US11374810B2 (en) 2017-11-03 2022-06-28 Vignet Incorporated Monitoring adherence and dynamically adjusting digital therapeutics
US11153156B2 (en) 2017-11-03 2021-10-19 Vignet Incorporated Achieving personalized outcomes with digital therapeutic applications
US11153159B2 (en) 2017-11-03 2021-10-19 Vignet Incorporated Digital therapeutics for precision medicine
US11381450B1 (en) 2017-11-03 2022-07-05 Vignet Incorporated Altering digital therapeutics over time to achieve desired outcomes
US20200119986A1 (en) * 2017-11-03 2020-04-16 Vignet Incorporated Systems and methods for managing operation of devices in complex systems and changing environments
US11700175B2 (en) * 2017-11-03 2023-07-11 Vignet Incorporated Personalized digital therapeutics to reduce medication side effects
US20210144058A1 (en) * 2017-11-03 2021-05-13 Vignet Incorporated Systems and methods for managing operation of devices in complex systems and changing environments
US20210327543A1 (en) * 2018-07-27 2021-10-21 Karydo Therapeutix, Inc. Artificial Intelligence Model for Predicting Actions of Test Substance in Humans
US11676684B2 (en) * 2018-07-27 2023-06-13 Karydo Therapeutix, Inc. Artificial intelligence model for predicting actions of test substance in humans
US11482305B2 (en) 2018-08-18 2022-10-25 Synkrino Biotherapeutics, Inc. Artificial intelligence analysis of RNA transcriptome for drug discovery
US11158423B2 (en) 2018-10-26 2021-10-26 Vignet Incorporated Adapted digital therapeutic plans based on biomarkers
US11923079B1 (en) 2019-02-01 2024-03-05 Vignet Incorporated Creating and testing digital bio-markers based on genetic and phenotypic data for therapeutic interventions and clinical trials
US11238979B1 (en) 2019-02-01 2022-02-01 Vignet Incorporated Digital biomarkers for health research, digital therapeautics, and precision medicine
WO2021211881A1 (en) * 2020-04-17 2021-10-21 Cipherome, Inc. Prediction of adverse drug reaction based on machine-learned models using protein function scores and clinical factors
US11302448B1 (en) 2020-08-05 2022-04-12 Vignet Incorporated Machine learning to select digital therapeutics
US11504011B1 (en) 2020-08-05 2022-11-22 Vignet Incorporated Early detection and prevention of infectious disease transmission using location data and geofencing
US11322260B1 (en) 2020-08-05 2022-05-03 Vignet Incorporated Using predictive models to predict disease onset and select pharmaceuticals
US11456080B1 (en) 2020-08-05 2022-09-27 Vignet Incorporated Adjusting disease data collection to provide high-quality health data to meet needs of different communities
US11789837B1 (en) 2021-02-03 2023-10-17 Vignet Incorporated Adaptive data collection in clinical trials to increase the likelihood of on-time completion of a trial
US11645180B1 (en) 2021-04-16 2023-05-09 Vignet Incorporated Predicting and increasing engagement for participants in decentralized clinical trials
US11281553B1 (en) 2021-04-16 2022-03-22 Vignet Incorporated Digital systems for enrolling participants in health research and decentralized clinical trials
US11586524B1 (en) 2021-04-16 2023-02-21 Vignet Incorporated Assisting researchers to identify opportunities for new sub-studies in digital health research and decentralized clinical trials
US11705230B1 (en) 2021-11-30 2023-07-18 Vignet Incorporated Assessing health risks using genetic, epigenetic, and phenotypic data sources
US11901083B1 (en) 2021-11-30 2024-02-13 Vignet Incorporated Using genetic and phenotypic data sets for drug discovery clinical trials
WO2023250312A1 (en) * 2022-06-21 2023-12-28 Parata Systems, Llc Methods, systems, and computer program product for pre-categorizing drug products based on characteristics thereof to reduce artificial intelligence model size used in validating the drug products

Also Published As

Publication number Publication date
US10262107B1 (en) 2019-04-16

Similar Documents

Publication Publication Date Title
US10262107B1 (en) Pharmacogenetic drug interaction management system
US11621083B2 (en) Cancer evolution detection and diagnostic
Staszak et al. Machine learning in drug design: Use of artificial intelligence to explore the chemical structure–biological activity relationship
Chen et al. The use of classification trees for bioinformatics
Gullapalli et al. Next generation sequencing in clinical medicine: Challenges and lessons for pathology and biomedical informatics
Singh et al. Integrative toxicogenomics: Advancing precision medicine and toxicology through artificial intelligence and OMICs technology
AU2015342771B2 (en) Predicting health outcomes
US20180060482A1 (en) Interpreting genomic results and providing targeted treatment options in cancer patients
CN107406876A (en) Show detection and treatment and the system and method for transmitting test result of the heterogeneous disease of sick cell
WO2018064547A1 (en) Methods for classifying somatic variations
Benard et al. Data mining for mutation-specific targets in acute myeloid leukemia
Magavern et al. The interface of therapeutics and genomics in cardiovascular medicine
US20210098075A1 (en) Method for managing test request by computer, management device, management computer program, and management system
Berlow Probabilistic Boolean modeling of pre‐clinical tumor models for biomarker identification in cancer drug development
Sikora Surrogate endpoints in cancer drug development
Yu et al. Genome-wide discovery of hidden genes mediating known drug-disease association using KDDANet
Dhara Computational Drug Repurposing for Breast Cancer Subtypes
Idda et al. Genetic variation among pharmacogenes in the Sardinian population
Shoket et al. Informatics and computational methods in natural product drug discovery
Allyn-Feuer The Pharmacoepigenomics Informatics Pipeline and H-GREEN Hi-C Compiler: Discovering Pharmacogenomic Variants and Pathways with the Epigenome and Spatial Genome
Sayer Application of Informatics Tools to Facilitate the Practice of Precision Medicine with Genomic Testing and Clinical Data
Hummel et al. Association between a prognostic gene signature and functional gene sets
Wang et al. DrugRepo: A novel approach to repurpose a huge collection of compounds based on chemical and genomic features
Qu Genomic Analysis of Micro-inversions Based on High-Throughput Sequencing
Shields Challenges and Opportunities in Clinical Bioinformatics

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION